301
|
Jiang X, Xu Y, Ren H, Jiang J, Wudu M, Wang Q, Guan J, Su H, Zhang Y, Zhang B, Guo Y, Hu Y, Jiang L, Liu Z, Wang H, Cheng Y, Sun L, Qiu X. KLHL18 inhibits the proliferation, migration, and invasion of non-small cell lung cancer by inhibiting PI3K/PD-L1 axis activity. Cell Biosci 2020; 10:139. [PMID: 33292627 PMCID: PMC7694932 DOI: 10.1186/s13578-020-00499-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Background The expression of Kelch-like protein 18 (KLHL18) in non-small cell lung cancer (NSCLC) is lower than that in normal lung tissue according to the Gene Expression Profiling Interactive Analysis database. KLHL18 is a BTB domain protein and binds cullin 3 (CUL3). However, whether this complex participates in ubiquitination-mediated protein degradation in NSCLC is unclear. Therefore, we aimed to investigate the role of KLHL18 in human NSCLC cells. Results We found that KLHL18 is downregulated in cancer cells and is associated with poor prognosis. Further, its expression was significantly associated with tumor node metastasis (TNM) stage, lymph node metastasis, and tumor size. In vitro analysis of NSCLC cells showed that overexpressing KLHL18 inhibited cell proliferation, migration, and invasion. We found that the tumor-inhibitory effect of the KLHL18 protein was achieved by promoting the ubiquitination and degradation of phosphatidylinositol 3-kinase (PI3K) p85α and inhibiting the expression of PD-L1 protein, ultimately preventing tumor cell immune escape. Conclusions Our results identified the tumor-suppressive mechanism of KLHL18 and suggested that it is closely related to NSCLC occurrence and development. Further investigation of the underlying mechanism may provide new targets for NSCLC treatment.
Collapse
Affiliation(s)
- Xizi Jiang
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yitong Xu
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Pathology, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning, People's Republic of China
| | - Hongjiu Ren
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Pathology, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning, People's Republic of China
| | - Jun Jiang
- Department of Pathology, The First Bethune Hospital of Jilin University, Changchun, Jilin, China
| | - Muli Wudu
- Department of Pathology, Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Qiongzi Wang
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jingqian Guan
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hongbo Su
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Pathology, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning, People's Republic of China
| | - Yao Zhang
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Bo Zhang
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yuanzi Guo
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yujiao Hu
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Lihong Jiang
- Department of Pathology, Chang'an District Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, No. 120 Wenyuan Middle Road, Guodu Street, Chang'an District, Xi'an, 710100, Shaanxi, China
| | - Zongang Liu
- Shengjing Hospital Affiliated With China Medical University, 19F, Building No. 1B, No. 36, Sanhao Street, Heping District, Shenyang, 110000, Liaoning, China
| | - Huanxi Wang
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yu Cheng
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Limei Sun
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Pathology, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning, People's Republic of China
| | - Xueshan Qiu
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China. .,Department of Pathology, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning, People's Republic of China.
| |
Collapse
|
302
|
Di Cintio F, Dal Bo M, Baboci L, De Mattia E, Polano M, Toffoli G. The Molecular and Microenvironmental Landscape of Glioblastomas: Implications for the Novel Treatment Choices. Front Neurosci 2020; 14:603647. [PMID: 33324155 PMCID: PMC7724040 DOI: 10.3389/fnins.2020.603647] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma (GBM) is the most frequent and aggressive primary central nervous system tumor. Surgery followed by radiotherapy and chemotherapy with alkylating agents constitutes standard first-line treatment of GBM. Complete resection of the GBM tumors is generally not possible given its high invasive features. Although this combination therapy can prolong survival, the prognosis is still poor due to several factors including chemoresistance. In recent years, a comprehensive characterization of the GBM-associated molecular signature has been performed. This has allowed the possibility to introduce a more personalized therapeutic approach for GBM, in which novel targeted therapies, including those employing tyrosine kinase inhibitors (TKIs), could be employed. The GBM tumor microenvironment (TME) exerts a key role in GBM tumor progression, in particular by providing an immunosuppressive state with low numbers of tumor-infiltrating lymphocytes (TILs) and other immune effector cell types that contributes to tumor proliferation and growth. The use of immune checkpoint inhibitors (ICIs) has been successfully introduced in numerous advanced cancers as well as promising results have been shown for the use of these antibodies in untreated brain metastases from melanoma and from non-small cell lung carcinoma (NSCLC). Consequently, the use of PD-1/PD-L1 inhibitors has also been proposed in several clinical trials for the treatment of GBM. In the present review, we will outline the main GBM molecular and TME aspects providing also the grounds for novel targeted therapies and immunotherapies using ICIs for GBM.
Collapse
Affiliation(s)
- Federica Di Cintio
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Lorena Baboci
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Maurizio Polano
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
303
|
Cai L, Duan J, Qian L, Wang Z, Wang S, Li S, Wang C, Zhao J, Zhang X, Bai H, Wang J. ROS1 Fusion Mediates Immunogenicity by Upregulation of PD-L1 After the Activation of ROS1-SHP2 Signaling Pathway in Non-Small Cell Lung Cancer. Front Immunol 2020; 11:527750. [PMID: 33324391 PMCID: PMC7723923 DOI: 10.3389/fimmu.2020.527750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022] Open
Abstract
The drug resistance of first-line crizotinib therapy for ROS proto-oncogene 1, receptor tyrosine kinase (ROS1) fusion non-small cell lung cancer (NSCLC) is inevitable. Whether the administration of immune checkpoint inhibitor (ICI) therapy is suitable for ROS 1 fusion NSCLCs or after the development of crizotinib resistance is still unknown. In this study, five different crizotinib resistant concentration cell lines (HCC78CR1-5) from primary sensitive HCC78 cells were cultured. Ba/F3 cells expressing crizotinib sensitive ROS1 fusion and crizotinib resistant ROS1-G2032R mutation were used to explore the relationship between ROS1 fusion, ROS1-G2032R mutation and programmed death-ligand 1 (PD-L1) expression and the clinical potential of anti-PD-L1 ICI therapy. The signaling pathway net was compared between HCC78 and HCC78CR1-5 cells using RNA sequencing. Anti- PD-L1 ICI therapy was performed on mouse xenograft models with Ba/F3 ROS1 fusion or ROS1-G2032R mutation. HCC78CR1-5 showed more immunogenicity than HCC78 in immune-related pathways. The PD-L1 expression level was remarkably higher in HCC78CR1-5 with ROS1 fusion upregulation than HCC78 primary cell. Furthermore, the expression of PD-L1 was down-regulated by RNA interference with ROS1 siRNAs and up-regulated lower in Ba/F3 ROS1-G2032R resistant mutation than ROS1 fusion. Western blotting analysis showed the ROS1-SHP2 signaling pathway activation in HCC78CR1-5 cells, Ba/F3 ROS1 fusion and ROS1-G2032R resistant mutation. Mouse xenograft models with Ba/F3 ROS1 fusion showed more CD3+PD-1+ T cells both in blood and tissue, and more sensitivity than the cells with Ba/F3 ROS1-G2032R resistant mutation after anti-PD-L1 therapy. Our findings indicate that PD-L1 upregulation depends on ROS1 fusion more than ROS1-G2032R mutation. We share our insights of NSCLCs treatment management into the use of anti-PD-L1 ICI therapy in ROS1 fusion and not in ROS1-G2032R resistant mutation.
Collapse
Affiliation(s)
- Liangliang Cai
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, China
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianchun Duan
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Qian
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, China
| | - Zhijie Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuhang Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sini Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Zhao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Zhang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua Bai
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
304
|
Singh SS, Dahal A, Shrestha L, Jois SD. Genotype Driven Therapy for Non-Small Cell Lung Cancer: Resistance, Pan Inhibitors and Immunotherapy. Curr Med Chem 2020; 27:5274-5316. [PMID: 30854949 DOI: 10.2174/0929867326666190222183219] [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: 10/05/2018] [Revised: 01/25/2019] [Accepted: 02/14/2019] [Indexed: 12/14/2022]
Abstract
Eighty-five percent of patients with lung cancer present with Non-small Cell Lung Cancer (NSCLC). Targeted therapy approaches are promising treatments for lung cancer. However, despite the development of targeted therapies using Tyrosine Kinase Inhibitors (TKI) as well as monoclonal antibodies, the five-year relative survival rate for lung cancer patients is still only 18%, and patients inevitably become resistant to therapy. Mutations in Kirsten Ras Sarcoma viral homolog (KRAS) and epidermal growth factor receptor (EGFR) are the two most common genetic events in lung adenocarcinoma; they account for 25% and 20% of cases, respectively. Anaplastic Lymphoma Kinase (ALK) is a transmembrane receptor tyrosine kinase, and ALK rearrangements are responsible for 3-7% of NSCLC, predominantly of the adenocarcinoma subtype, and occur in a mutually exclusive manner with KRAS and EGFR mutations. Among drug-resistant NSCLC patients, nearly half exhibit the T790M mutation in exon 20 of EGFR. This review focuses on some basic aspects of molecules involved in NSCLC, the development of resistance to treatments in NSCLC, and advances in lung cancer therapy in the past ten years. Some recent developments such as PD-1-PD-L1 checkpoint-based immunotherapy for NSCLC are also covered.
Collapse
Affiliation(s)
- Sitanshu S Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201, United States
| | - Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201, United States
| | - Leeza Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201, United States
| | - Seetharama D Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201, United States
| |
Collapse
|
305
|
Gou Q, Dong C, Xu H, Khan B, Jin J, Liu Q, Shi J, Hou Y. PD-L1 degradation pathway and immunotherapy for cancer. Cell Death Dis 2020; 11:955. [PMID: 33159034 PMCID: PMC7648632 DOI: 10.1038/s41419-020-03140-2] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/20/2022]
Abstract
Programmed death ligand 1 (PD-L1, CD274) is an essential immune checkpoint protein that binds to programmed death 1 (PD-1) on T-lymphocytes. T cell plays a critical role in killing cancer cells while the cancer cell exhibits immune escape by the expression of PD-L1. The binding of PD-L1 to PD-1 inhibits T cell proliferation and activity, leading to tumor immunosuppression. Increasing evidence shows that PD-L1 protein undergoes degradation in proteasomes or lysosomes by multiple pathways, leading to enhanced immunotherapy for cancer. Although some specific drugs induce PD-L1 degradation and increase antitumor activity, the combination of these drugs with PD-L1/PD-1 blockade significantly enhances cancer immunotherapy. In this review, we have discussed the interaction of PD-L1 degradation with cancer immunotherapy.
Collapse
Affiliation(s)
- Qian Gou
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University, Changzhou, Jiangsu Province, 213017, China.,School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China.,School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Chen Dong
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Huihui Xu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Bibimaryam Khan
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Jianhua Jin
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University, Changzhou, Jiangsu Province, 213017, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Xuzhou, Jiangsu Province, 212017, China
| | - Qian Liu
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University, Changzhou, Jiangsu Province, 213017, China.,Department of Oncology, The Wujin Clinical College of Xuzhou Medical University, Xuzhou, Jiangsu Province, 212017, China
| | - Juanjuan Shi
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| | - Yongzhong Hou
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University, Changzhou, Jiangsu Province, 213017, China. .,School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China.
| |
Collapse
|
306
|
Yanagawa N, Shiono S, Endo M, Ogata SY, Yamada N, Sugimoto R, Osakabe M, Uesugi N, Sugai T. Programmed death ligand 1 protein expression is positively correlated with the solid predominant subtype, high MIB-1 labeling index, and p53 expression and negatively correlated with epidermal growth factor receptor mutations in lung adenocarcinoma. Hum Pathol 2020; 108:12-21. [PMID: 33159965 DOI: 10.1016/j.humpath.2020.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/24/2020] [Accepted: 10/30/2020] [Indexed: 12/24/2022]
Abstract
Programmed death ligand 1 (PD-L1) protein expression is a proposed predictive biomarker of immunotherapy; thus, identification of the clinicopathological and molecular characteristics associated with PD-L1 expression is important and necessary. We examined PD-L1 immunohistochemical expression and its relationships with the clinicopathological and molecular characteristics of patients with surgically resected nonsmall cell lung carcinoma. PD-L1 expression differed according to the histological subtype. Among 633 patients with adenocarcinoma, 523 (82.6%) had no PD-L1 expression, 78 (12.3%) low expression, and 32 (5.1%) high expression. PD-L1 expression was more common in men (p < 0.001), in smokers (p = 0.002), and in patients with a more advanced stage (p = 0.002), the solid predominant subtype (p < 0.001), no epidermal growth factor receptor(EGFR) mutations (p < 0.001), a high MIB-1 labeling index (p < 0.001), and positive p53 immunohistochemical expression (p < 0.001). In a multivariate logistic regression analysis, the solid predominant subtype (odds ratio [OR] = 4.92, 95% confidence interval [CI]: 2.72-8.89, p < 0.001), no EGFR mutations (OR = 2.27, 95% CI: 1.35-2.7, p = 0.002), a high MIB-1 labeling index (OR = 2.78, 95% CI: 1.72-4.55, p < 0.001), and p53 positivity (OR = 2.13, 95% CI: 1.34-4.36, p = 0.042) were significantly and independently associated with PD-L1 expression. The combination of the solid predominant subtype with a high MIB-1 labeling index was strongly associated with positive expression of PD-L1. In the 193 patients with squamous cell carcinoma, 92 (47.7%) had no PD-L1 expression, 57 (29.5%) low expression, and 44 (22.8%) high expression. There were no significant correlations between PD-L1 expression and the evaluated clinicopathological or molecular characteristics of these patients. These results, indicating associations of PD-L1 with various clinicopathological or molecular characteristics in adenocarcinoma but not squamous cell carcinoma, may be useful for selecting patients with a good response to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Naoki Yanagawa
- Department of Molecular Diagnostic Pathology, Iwate Medical University, Shiwa-gun, Iwate, 0283695, Japan.
| | - Satoshi Shiono
- Departments of Thoracic Surgery and Yamagata Prefectural Central Hospital, Yamagata, Yamagata, 9902292, Japan
| | - Makoto Endo
- Departments of Thoracic Surgery and Yamagata Prefectural Central Hospital, Yamagata, Yamagata, 9902292, Japan
| | - Shin-Ya Ogata
- Diagnostic Pathology, Yamagata Prefectural Central Hospital, Yamagata, Yamagata, 9902292, Japan
| | - Noriyuki Yamada
- Department of Molecular Diagnostic Pathology, Iwate Medical University, Shiwa-gun, Iwate, 0283695, Japan
| | - Ryo Sugimoto
- Department of Molecular Diagnostic Pathology, Iwate Medical University, Shiwa-gun, Iwate, 0283695, Japan
| | - Mitsumasa Osakabe
- Department of Molecular Diagnostic Pathology, Iwate Medical University, Shiwa-gun, Iwate, 0283695, Japan
| | - Noriyuki Uesugi
- Department of Molecular Diagnostic Pathology, Iwate Medical University, Shiwa-gun, Iwate, 0283695, Japan
| | - Tamotsu Sugai
- Department of Molecular Diagnostic Pathology, Iwate Medical University, Shiwa-gun, Iwate, 0283695, Japan
| |
Collapse
|
307
|
Glorieux C, Xia X, He YQ, Hu Y, Cremer K, Robert A, Liu J, Wang F, Ling J, Chiao PJ, Huang P. Regulation of PD-L1 expression in K-ras-driven cancers through ROS-mediated FGFR1 signaling. Redox Biol 2020; 38:101780. [PMID: 33171331 PMCID: PMC7658718 DOI: 10.1016/j.redox.2020.101780] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/31/2022] Open
Abstract
K-ras mutations are major genetic events that drive cancer development associated with aggressive malignant phenotypes, while expression of the immune checkpoint molecule PD-L1 plays a key role in cancer evasion of the immune surveillance that also profoundly affects the patient outcome. However, the relationship between K-ras oncogenic signal and PD-L1 expressions as an important area that requires further investigation. Using both in vitro and in vivo experimental models of K-ras-driven cancer, we found that oncogenic K-ras significantly enhanced PD-L1 expression through a redox-mediated mechanism. Activation of K-rasG12V promoted ROS generation and induced FGFR1 expression, leading to a significant upregulation of PD-L1. We further showed that exogenous ROS such as hydrogen peroxide alone was sufficient to activate FGFR1 and induce PD-L1, while antioxidants could largely abrogate PD-L1 expression in K-ras mutant cells, indicating a critical role of redox regulation. Importantly, genetic knockout of FGFR1 led to a decrease in PD-L1 expression, and impaired tumor growth in vivo due to a significant increase of T cell infiltration in the tumor tissues and thus enhanced T-cell-mediated tumor suppression. Our study has identified a novel mechanism by which K-ras promotes PD-L1 expression, and suggests that modulation of ROS or inhibition of the FGFR1 pathway could be a novel strategy to abrogate PD-L1-mediated immunosuppression and thus potentially improve the efficacy of immunotherapy in K-ras-driven cancers. Oncogenic K-Ras up-regulates PD-L1 expression in vitro and in vivo. ROS play a major role in mediating K-Ras-induced FGFR1 activation leading to PD-L1 expression in K-Ras-driven cancers. Antioxidants are able to modulate PD-L1 expression in K-Ras mutant cancer cells. Suppression of FGFR1 enhances CD8+ T cell infiltration and inhibits tumor growth.
Collapse
Affiliation(s)
- Christophe Glorieux
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
| | - Xiaojun Xia
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yong-Qiao He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yumin Hu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Kelly Cremer
- Pôle Epidémiologie et Biostatistique, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, 1200, Belgium
| | - Annie Robert
- Pôle Epidémiologie et Biostatistique, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, 1200, Belgium
| | - Junchen Liu
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston TX, 77030, Texas, USA
| | - Fen Wang
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston TX, 77030, Texas, USA
| | - Jianhua Ling
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston TX, 77030, Texas, USA
| | - Paul J Chiao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston TX, 77030, Texas, USA
| | - Peng Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
| |
Collapse
|
308
|
Zhang L, Zhang M, Xu J, Li S, Chen Y, Wang W, Yang J, Li S, Gu M. The role of the programmed cell death protein-1/programmed death-ligand 1 pathway, regulatory T cells and T helper 17 cells in tumor immunity: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1526. [PMID: 33313271 PMCID: PMC7729304 DOI: 10.21037/atm-20-6719] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tumor immunotherapy, especially that involving programmed cell death protein-1 (PD-1)/programmed death-ligand 1 (PD-L1) immunosuppressive checkpoint inhibitors, has become an important part of tumor treatment strategy in the past decade. Blocking PD-1/PD-L1 signaling pathway can reduce the inhibitory effect of PD-1 pathway on T cells, promote the anti-tumor activity of activated T cells, and prolong the remission period of tumor. While PD-1/PD-L1 immunotherapy is effective in the treatment of solid malignant tumors, it also has shortcomings, due to the complexity of the tumor microenvironment (TME). Regulatory T cells (Tregs) and T helper 17 (Th17) cells play an important role in the TME and are closely related to the occurrence and development of tumors. Tregs can inhibit the anti-tumor immune effect, while Th17 cells play a dual role in tumor immunity, which not only promotes tumorigenesis but also promotes anti-tumor immunity. In the occurrence and development of tumor, PD-1/PD-L1 pathway, Tregs and Th17 cells are interrelated. However, the complicated relationship between the PD-1/PD-L1 pathway, Tregs, and Th17 cells has not been fully clarified. Here, we summarize the immunoregulation mechanisms and discuss the crosstalk between the PD-1/PD-L1 pathway, Tregs, and Th17 cells, with the aim of providing novel insights for future cancer treatment.
Collapse
Affiliation(s)
- Lanfang Zhang
- Department of Chemotherapy Unit 2, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Mingjuan Zhang
- Department of Chemotherapy Unit 2, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Jinxiu Xu
- Department of Chemotherapy Unit 2, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Shan Li
- Department of Chemotherapy Unit 2, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Yu Chen
- Department of Chemotherapy Unit 2, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Wenjing Wang
- Department of Chemotherapy Unit 2, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Juntian Yang
- Department of Chemotherapy Unit 2, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Shengyun Li
- Department of Chemotherapy Unit 2, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Meiling Gu
- Department of Chemotherapy Unit 2, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| |
Collapse
|
309
|
Xu J, Nie H, He J, Wang X, Liao K, Tu L, Xiong Z. Using Machine Learning Modeling to Explore New Immune-Related Prognostic Markers in Non-Small Cell Lung Cancer. Front Oncol 2020; 10:550002. [PMID: 33215029 PMCID: PMC7665579 DOI: 10.3389/fonc.2020.550002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To find new immune-related prognostic markers for non-small cell lung cancer (NSCLC). METHODS We found GSE14814 is related to NSCLC in GEO database. The non-small cell lung cancer observation (NSCLC-OBS) group was evaluated for immunity and divided into high and low groups for differential gene screening according to the score of immune evaluation. A single factor COX regression analysis was performed to select the genes related to prognosis. A prognostic model was constructed by machine learning, and test whether the model has a test efficacy for prognosis. A chip-in-chip non-small cell lung cancer chemotherapy (NSCLC-ACT) sample was used as a validation dataset for the same validation and prognostic analysis of the model. The coexpression genes of hub genes were obtained by pearson analysis and gene enrichment, function enrichment and protein interaction analysis. The tumor samples of patients with different clinical stages were detected by immunohistochemistry and the expression difference of prognostic genes in tumor tissues of patients with different stages was compared. RESULTS By screening, we found that LYN, C3, COPG2IT1, HLA.DQA1, and TNFRSF17 is closely related to prognosis. After machine learning, we constructed the immune prognosis model from these 5 genes, and the model AUC values were greater than 0.9 at three time periods of 1, 3, and 5 years; the total survival period of the low-risk group was significantly better than that of the high-risk group. The results of prognosis analysis in ACT samples were consistent with OBS groups. The coexpression genes are mainly involved B cell receptor signaling pathway and are mainly enriched in apoptotic cell clearance. Prognostic key genes are highly correlated with PDCD1, PDCD1LG2, LAG3, and CTLA4 immune checkpoints. The immunohistochemical results showed that the expression of COPG2IT1 and HLA.DQA1 in stage III increased significantly and the expression of LYN, C3, and TNFRSF17 in stage III decreased significantly compared with that of stage I. The experimental results are consistent with the previous analysis. CONCLUSION LYN, C3, COPG2IT1, LA.DQA1, and NFRSF17 may be new immune markers to judge the prognosis of patients with non-small cell lung cancer.
Collapse
Affiliation(s)
- Jiasheng Xu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Han Nie
- Department of Vascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiarui He
- Department of Vascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xinlu Wang
- Department of Vascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kaili Liao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Luxia Tu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenfang Xiong
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
310
|
Hamarsheh S, Groß O, Brummer T, Zeiser R. Immune modulatory effects of oncogenic KRAS in cancer. Nat Commun 2020; 11:5439. [PMID: 33116132 PMCID: PMC7595113 DOI: 10.1038/s41467-020-19288-6] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023] Open
Abstract
Oncogenic KRAS mutations are the most frequent mutations in human cancer, but most difficult to target. While sustained proliferation caused by oncogenic KRAS-downstream signalling is a main driver of carcinogenesis, there is increasing evidence that it also mediates autocrine effects and crosstalk with the tumour microenvironment (TME). Here, we discuss recent reports connecting KRAS mutations with tumour-promoting inflammation and immune modulation caused by KRAS that leads to immune escape in the TME. We discuss the preclinical work on KRAS-induced inflammation and immune modulation in the context of currently ongoing clinical trials targeting cancer entities that carry KRAS mutations and strategies to overcome the oncogene-induced effects on the immune system.
Collapse
Affiliation(s)
- Shaima'a Hamarsheh
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Olaf Groß
- Institute of Neuropathology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Centre for Biological Signalling Studies (BIOSS) and Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Centre Freiburg (CCCF), University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Centre for Biological Signalling Studies (BIOSS) and Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany. .,German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Comprehensive Cancer Centre Freiburg (CCCF), University of Freiburg, Freiburg, Germany.
| |
Collapse
|
311
|
Lamberti G, Sisi M, Andrini E, Palladini A, Giunchi F, Lollini PL, Ardizzoni A, Gelsomino F. The Mechanisms of PD-L1 Regulation in Non-Small-Cell Lung Cancer (NSCLC): Which Are the Involved Players? Cancers (Basel) 2020; 12:E3129. [PMID: 33114576 PMCID: PMC7692442 DOI: 10.3390/cancers12113129] [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: 09/16/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/27/2022] Open
Abstract
Treatment with inhibition of programmed cell death 1 (PD-1) or its ligand (PD-L1) improves survival in advanced non-small-cell lung cancer (NSCLC). Nevertheless, only a subset of patients benefit from treatment and biomarkers of response to immunotherapy are lacking. Expression of PD-L1 on tumor cells is the primary clinically-available predictive factor of response to immune checkpoint inhibitors, and its relevance in cancer immunotherapy has fostered several studies to better characterize the mechanisms that regulate PD-L1 expression. However, the factors associated with PD-L1 expression are still not well understood. Genomic alterations that activate KRAS, EGFR, and ALK, as well as the loss of PTEN, have been associated with increased PD-L1 expression. In addition, PD-L1 expression is reported to be increased by amplification of CD274, and decreased by STK11 deficiency. Furthermore, PD-L1 expression can be modulated by either tumor extrinsic or intrinsic factors. Among extrinsic factors, the most prominent one is interferon-γ release by immune cells, while there are several tumor intrinsic factors such as activation of the mechanistic target of rapamycin (mTOR), mitogen-activated protein kinase (MAPK) and Myc pathways that can increase PD-L1 expression. A deeper understanding of PD-L1 expression regulation is crucial for improving strategies that exploit inhibition of this immune checkpoint in the clinic, especially in NSCLC where it is central in the therapeutic algorithm. We reviewed current preclinical and clinical data about PD-L1 expression regulation in NSCLC.
Collapse
Affiliation(s)
- Giuseppe Lamberti
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Alma Mater Studiorum University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.L.); (M.S.); (E.A.); (A.A.)
| | - Monia Sisi
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Alma Mater Studiorum University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.L.); (M.S.); (E.A.); (A.A.)
| | - Elisa Andrini
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Alma Mater Studiorum University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.L.); (M.S.); (E.A.); (A.A.)
| | - Arianna Palladini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, viale Filopanti 22, 40126 Bologna, Italy; (A.P.); (P.-L.L.)
| | - Francesca Giunchi
- Laboratory of Oncologic Molecular Pathology, S.Orsola-Malpighi Teaching Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, viale Filopanti 22, 40126 Bologna, Italy; (A.P.); (P.-L.L.)
| | - Andrea Ardizzoni
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Alma Mater Studiorum University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.L.); (M.S.); (E.A.); (A.A.)
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy
| | - Francesco Gelsomino
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy
| |
Collapse
|
312
|
Hua P, Zhang Y, Jin C, Zhang G, Wang B. Integration of gene profile to explore the hub genes of lung adenocarcinoma: A quasi-experimental study. Medicine (Baltimore) 2020; 99:e22727. [PMID: 33120770 PMCID: PMC7581154 DOI: 10.1097/md.0000000000022727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Lung cancer is a leading cause of morbidity diseases worldwide, but the key mechanisms of lung cancer remain elusive. This study aims to integrate of GSE 118370 and GSE 32863 profile and identify the key genes and pathway involved in human lung adenocarcinoma. METHODS R software (RStudio, Version info: R 3.2.3, Forrester, USA) were utilized to find the differentially expressed genes. All the differentially expressed genes were analyzed by gene ontology, kyoto encyclopedia of genes and genomes. Protein-protein interaction networks were constructed by STRING database and analyzed by Cytohubber and Module. The cancer genome atlas database was used to verification the expression of hub genes. Quantitative reverse transcription-PCR was used to verify the bio-information results. RESULTS Sixty-four lung adenocarcinoma and 64 adjacent normal tissues were used for integration analysis. Five hundred ninety-nine co-expression genes were locked. Biological processes mainly enriched in angiogenesis. Cellular component focused on extracellular exosome and molecular function aimed on protein disulfide isomerase activity. Cytohubber analysis showed that GNG11, FPR2, P4HB, PIK3R1, CDC20, ADCY4, TIMP1, IL6, CXC chemokine ligand (CXCL)12, and GAS6 acted as the hub genes during lung adenocarcinoma. Module analysis presented Chemokine signaling pathway was a key pathway. Quantitative reverse transcription-PCR showed that the expression level of GNG11, FPR2, PIK3R1, ADCY4, IL6, CXCL12, and GAS6 were significantly decreased and P4HB, CDC20 and TIMP1 were increased in human adenocarcinoma tissues (P < .05). The cancer genome atlas online analysis showed GNG11 was not associated with survival. CONCLUSIONS This study firstly reported GNG11 acting as a hub gene in adenocarcinoma. GNG11 could be used as a biomarker for human adenocarcinoma. Chemokine signaling pathway might play important roles in lung adenocarcinoma.
Collapse
|
313
|
Hudson K, Cross N, Jordan-Mahy N, Leyland R. The Extrinsic and Intrinsic Roles of PD-L1 and Its Receptor PD-1: Implications for Immunotherapy Treatment. Front Immunol 2020; 11:568931. [PMID: 33193345 PMCID: PMC7609400 DOI: 10.3389/fimmu.2020.568931] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022] Open
Abstract
Programmed death-ligand 1 (PD-L1) is an immune checkpoint inhibitor that binds to its receptor PD-1 expressed by T cells and other immune cells to regulate immune responses; ultimately preventing exacerbated activation and autoimmunity. Many tumors exploit this mechanism by overexpressing PD-L1 which often correlates with poor prognosis. Some tumors have also recently been shown to express PD-1. On tumors, PD-L1 binding to PD-1 on immune cells promotes immune evasion and tumor progression, primarily by inhibition of cytotoxic T lymphocyte effector function. PD-1/PD-L1-targeted therapy has revolutionized the cancer therapy landscape and has become the first-line treatment for some cancers, due to their ability to promote durable anti-tumor immune responses in select patients with advanced cancers. Despite this clinical success, some patients have shown to be unresponsive, hyperprogressive or develop resistance to PD-1/PD-L1-targeted therapy. The exact mechanisms for this are still unclear. This review will discuss the current status of PD-1/PD-L1-targeted therapy, oncogenic expression of PD-L1, the new and emerging tumor-intrinisic roles of PD-L1 and its receptor PD-1 and how they may contribute to tumor progression and immunotherapy responses as shown in different oncology models.
Collapse
Affiliation(s)
| | | | | | - Rebecca Leyland
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| |
Collapse
|
314
|
Li TE, Wang S, Shen XT, Zhang Z, Chen M, Wang H, Zhu Y, Xu D, Hu BY, Wei R, Zheng Y, Dong QZ, Qin LX. PKM2 Drives Hepatocellular Carcinoma Progression by Inducing Immunosuppressive Microenvironment. Front Immunol 2020; 11:589997. [PMID: 33193421 PMCID: PMC7606949 DOI: 10.3389/fimmu.2020.589997] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/29/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND AIMS Pyruvate kinase M2 (PKM2) is an essential regulator of the Warburg effect, but its biological function promoting immune escape of hepatocellular carcinoma (HCC) is unclear. METHODS GEPIA web tool and immunohistochemistry (IHC) analysis were employed to evaluate the clinical relevance of PKM2 in HCC patients. Both in vitro CCK-8, colony formation, and transwell assays, and in vivo xenografts were performed to evaluate the malignancy of HCC cells. PKM2 and PD-L1 levels were examined by Western blot, qRT-PCR, and IHC. The role of PKM2 on in vivo immune response was also investigated. RESULTS PKM2 was significantly upregulated in HCC and associated with a poor prognosis of HCC patients. Knockdown of PKM2 inhibited in vitro proliferation, migration, and invasion of HCC cells, as well as in vivo tumor growth. Strikingly, PKM2 showed a strong correlation with the expression of immune inhibitory cytokines and lymphocyte infiltration in HCC. The overexpression of PKM2 sensitized HCC to immune checkpoint blockade, which enhanced IFN-γ positive CD8 T cells in HCC mice models. CONCLUSION PKM2 might be a predictor and a potential therapeutic target for immune checkpoint inhibitors in HCC.
Collapse
Affiliation(s)
- Tian-En Li
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Shun Wang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xiao-Tian Shen
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Ze Zhang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Mo Chen
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Hao Wang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Ying Zhu
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Da Xu
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Bei-Yuan Hu
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Ran Wei
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Yan Zheng
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Qiong-Zhu Dong
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
315
|
Wang F, Wang S, Zhou Q. The Resistance Mechanisms of Lung Cancer Immunotherapy. Front Oncol 2020; 10:568059. [PMID: 33194652 PMCID: PMC7606919 DOI: 10.3389/fonc.2020.568059] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy has revolutionized lung cancer treatment in the past decade. By reactivating the host’s immune system, immunotherapy significantly prolongs survival in some advanced lung cancer patients. However, resistance to immunotherapy is frequent, which manifests as a lack of initial response or clinical benefit to therapy (primary resistance) or tumor progression after the initial period of response (acquired resistance). Overcoming immunotherapy resistance is challenging owing to the complex and dynamic interplay among malignant cells and the defense system. This review aims to discuss the mechanisms that drive immunotherapy resistance and the innovative strategies implemented to overcome it in lung cancer.
Collapse
Affiliation(s)
- Fen Wang
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangdong Lung Cancer Institute, South China University of Technology, Guangzhou, China.,Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Peking University Shenzhen Hospital, Shenzhen, China
| | - Shubin Wang
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qing Zhou
- Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, Guangdong Lung Cancer Institute, South China University of Technology, Guangzhou, China
| |
Collapse
|
316
|
Sinha D, Smith C, Khanna R. Joining Forces: Improving Clinical Response to Cellular Immunotherapies with Small-Molecule Inhibitors. Trends Mol Med 2020; 27:75-90. [PMID: 33011081 DOI: 10.1016/j.molmed.2020.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 07/24/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
Adoptive T cell therapy (ACT) has emerged as a powerful therapeutic tool against both hematological and virus-associated cancers. However, extension of this success to solid cancers has been challenging owing to intratumoral mechanisms that induce a hostile immunosuppressive tumor microenvironment (TME). Delineating the impact of tumor-intrinsic adaptive resistance mechanisms on immune-based therapies is essential to improve long-term efficacy. We discuss the different tumor-intrinsic factors that lead to resistance to ACT. We highlight the potential of repurposing molecular targeted therapies to modulate immune responses and override intratumor resistance to ACT. Finally, we discuss the potential of combining targeted therapy and ACT as a new paradigm to improve the clinical efficacy of cancer therapeutics.
Collapse
Affiliation(s)
- Debottam Sinha
- QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
| | - Corey Smith
- QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Medicine, University of Queensland, Brisbane, QLD, Australia.
| | - Rajiv Khanna
- QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Medicine, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
317
|
Spatial and Temporal Changes in PD-L1 Expression in Cancer: The Role of Genetic Drivers, Tumor Microenvironment and Resistance to Therapy. Int J Mol Sci 2020; 21:ijms21197139. [PMID: 32992658 PMCID: PMC7583014 DOI: 10.3390/ijms21197139] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Immunotherapies blocking immune inhibitory receptors programmed cell death-1 (PD-1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) on T-cells have dramatically improved patient outcomes in a range of advanced cancers. However, the lack of response, and the development of resistance remain major obstacles to long-term improvements in patient outcomes. There is significant interest in the clinical use of biomarkers to improve patient selection, and the expression of PD-1 ligand 1 (PD-L1) is often reported as a potential biomarker of response. However, accumulating evidence suggests that the predictive value of PD-L1 expression in tumor biopsies is relatively low due, in part, to its complex biology. In this review, we discuss the biological consequences of PD-L1 expression by various cell types within the tumor microenvironment, and the complex mechanisms that regulate PD-L1 expression at the genomic, transcriptomic and proteomic levels.
Collapse
|
318
|
Wang H, Zhang Z, Yan Z, Ma S. CKS1B promotes cell proliferation and invasion by activating STAT3/PD-L1 and phosphorylation of Akt signaling in papillary thyroid carcinoma. J Clin Lab Anal 2020; 35:e23565. [PMID: 32960462 PMCID: PMC7843292 DOI: 10.1002/jcla.23565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/05/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
Objective To investigate role of GKS1B and its relationship between STAT3/PD‐L1 and p‐Akt in papillary thyroid carcinoma (PTC). Methods Expression of GKS1B and PD‐L1 was determined in PTC cell lines. GKS1B was overexpressed or knocked down by transfection with overexpression plasmids or si‐CKS1B. STAT3 inhibitor WP1066 was used to suppress STAT3, and PD‐L1 inhibitor Pembrolizumab was used to block PD‐L1. Cell viability and invasion were evaluated by MTT and transwell assay, respectively. The expression of STAT3, p‐STAT3, Akt, and p‐Akt was measured using Western blotting. Results Both protein levels and mRNA levels of CKS1B and PD‐L1 were remarkably up‐regulated in PTC cell lines. Knockdown of CKS1B significantly inhibited cell viability and invasion of PTC cells and suppressed STAT3/PD‐L1 signaling and Akt phosphorylation, while overexpression of CKS1B led to opposite results. Inhibition of STAT3 or PD‐L1 reversed the effects of overexpressed CKS1B on PTC cells. Conclusion The overexpression of CSK1B could promote cell viability and invasion of PTC cells through activation of STAT3/PD‐L1 signaling and Akt phosphorylation.
Collapse
Affiliation(s)
- Hui Wang
- Shanghai Xuhui Center Hospital, Shanghai, China
| | | | - Zhe Yan
- Shanghai Xuhui Center Hospital, Shanghai, China
| | - Shihong Ma
- Shanghai Xuhui Center Hospital, Shanghai, China
| |
Collapse
|
319
|
Seol HY, Kim YS, Kim SJ. Predictive value of 18F-fluorodeoxyglucose positron emission tomography/computed tomography for PD-L1 expression in non-small cell lung cancer: A systematic review and meta-analysis. Thorac Cancer 2020; 11:3260-3268. [PMID: 32951338 PMCID: PMC7605997 DOI: 10.1111/1759-7714.13664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 12/26/2022] Open
Abstract
Background The purpose of the current study was to investigate the predictive value of 18F‐fluorodeoxyglucose positron emission tomography/computed tomography (18F‐FDG PET/CT) for programmed death ligand 1 (PD‐L1) in non‐small cell lung cancer (NSCLC) patients through a systematic review and meta‐analysis. Methods The PubMed, Cochrane, and EMBASE database, from the earliest available date of indexing through 30 April 2020, were searched for studies evaluating the diagnostic performance of 18F‐FDG PET/CT for prediction of PD‐L1 expression in NSCLC patients. Results Across six studies (1739 patients), the pooled sensitivity for 18F‐FDG PET/CT was 0.72 (95% CI: 0.58–0.82) with heterogeneity (I2 = 90.9, P < 0.001) and a pooled specificity of 0.69 (95% CI: 0.64–0.74) with heterogeneity (I2 = 77.9, P < 0.001). Likelihood ratio (LR) syntheses gave an overall positive likelihood ratio (LR +) of 2.3 (95% CI: 1.8–2.9) and negative likelihood ratio (LR‐) of 0.41 (95% CI: 0.26–0.63). The pooled diagnostic odds ratio (DOR) was six (95% CI: 3–11). Hierarchical summary receiver operating characteristic (ROC) curve indicated that the area under the curve was 0.74 (95% CI: 0.70–0.78). Conclusions The current meta‐analysis showed a moderate sensitivity and specificity of 18F‐FDG PET/CT for the prediction of PD‐L1 expression in NSCLC patients. The DOR was low and the likelihood ratio scatter‐gram indicated that 18F‐FDG PET/CT might not be useful for the prediction of PD‐L1 expression in NSCLC patients and not for its exclusion. Key points Significant findings of the study The current meta‐analysis showed a moderate sensitivity and specificity of 18F‐FDG PET/CT for the prediction of PD‐L1 expression in NSCLC patients. The DOR was low and the likelihood ratio scattergram indicated that 18F‐FDG PET/CT might not be useful for the prediction of PD‐L1 expression in NSCLC patients and not for its exclusion. What this study adds This study concluded that the role of 18F‐FDG PET/CT in predicting tumor expression of PD‐L1 should be further elucidated.
Collapse
Affiliation(s)
- Hee Yun Seol
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Yun Seong Kim
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Seong-Jang Kim
- Department of Nuclear Medicine, College of Medicine, Pusan National University, Yangsan, South Korea.,Department of Nuclear Medicine, Pusan National University Yangsan Hospital, Yangsan, South Korea.,BioMedical Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| |
Collapse
|
320
|
Yang H, Heyer J, Zhao H, Liang S, Guo R, Zhong L. The Potential Role of Cathepsin K in Non-Small Cell Lung Cancer. Molecules 2020; 25:molecules25184136. [PMID: 32927648 PMCID: PMC7571067 DOI: 10.3390/molecules25184136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/25/2022] Open
Abstract
(1) Background: Cathepsin K has been found overexpressed in several malignant tumors. However, there is little information regarding the involvement of Cathepsin K in non-small cell lung cancer (NSCLC). (2) Methods: Cathepsin K expression was tested in human NSCLC cell lines A549 and human embryo lung fibroblast MRC-5 cells using Western blot and immunofluorescence assay. Cathepsin K was transiently overexpressed or knocked down using transfection with a recombinant plasmid and siRNA, respectively, to test the effects on cell proliferation, migration, invasion, and on the mammalian target of rapamycin (mTOR) signaling pathway. (3) Results: Expression of Cathepsin K was increased significantly in A549 cells and diffused within the cytoplasm compared to the MRC-5 cells used as control. Cathepsin K overexpression promoted the proliferation, migration, and invasion of A549 cells, accompanied by mTOR activation. Cathepsin K knockdown reversed the above malignant behavior and inhibited the mTOR signaling activation, suggesting that Cathepsin K may promote the progression of NSCLC by activating the mTOR signaling pathway. (4) Conclusion: Cathepsin K may potentially represent a viable drug target for NSCLC treatment.
Collapse
Affiliation(s)
- Hui Yang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071000, China; (H.Y.); (H.Z.); (S.L.)
| | - Jasmine Heyer
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Hui Zhao
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071000, China; (H.Y.); (H.Z.); (S.L.)
| | - Shengxian Liang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071000, China; (H.Y.); (H.Z.); (S.L.)
| | - Rui Guo
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071000, China; (H.Y.); (H.Z.); (S.L.)
- Correspondence: (R.G.); (L.Z.)
| | - Li Zhong
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071000, China; (H.Y.); (H.Z.); (S.L.)
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA;
- Correspondence: (R.G.); (L.Z.)
| |
Collapse
|
321
|
Wang L, Gao Y, Zhang G, Li D, Wang Z, Zhang J, Hermida LC, He L, Wang Z, Si J, Geng S, Ai R, Ning F, Cheng C, Deng H, Dimitrov DS, Sun Y, Huang Y, Wang D, Hu X, Wei Z, Wang W, Liao X. Enhancing KDM5A and TLR activity improves the response to immune checkpoint blockade. Sci Transl Med 2020; 12. [DOI: 10.1126/scitranslmed.aax2282] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
The bifunctional compound D18 improves checkpoint blockade efficacy by increasing KDM5A and PD-L1 abundance and inducing TLR7/8 activation.
Collapse
Affiliation(s)
- Liangliang Wang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Beijing Advanced Innovation Center for Human Brain Protection, Tsinghua University, Beijing 100084, China
| | - Yan Gao
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Beijing Advanced Innovation Center for Human Brain Protection, Tsinghua University, Beijing 100084, China
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, China
| | - Gao Zhang
- Department of Neurosurgery and The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Dan Li
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Beijing Advanced Innovation Center for Human Brain Protection, Tsinghua University, Beijing 100084, China
| | - Zhenda Wang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Beijing Advanced Innovation Center for Human Brain Protection, Tsinghua University, Beijing 100084, China
| | - Jie Zhang
- Department of Computer Science, College of Computing Sciences, New Jersey Institute of Technology, Neswark, NJ 07102, USA
| | - Leandro C. Hermida
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Center for Bioinformatics and Computational Biology, Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | - Lei He
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Beijing Advanced Innovation Center for Human Brain Protection, Tsinghua University, Beijing 100084, China
| | - Zhisong Wang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Beijing Advanced Innovation Center for Human Brain Protection, Tsinghua University, Beijing 100084, China
| | - Jingwen Si
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Beijing Advanced Innovation Center for Human Brain Protection, Tsinghua University, Beijing 100084, China
| | - Shuang Geng
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics (ICG), College of Chemistry, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Rizi Ai
- Department of Chemistry and Biochemistry, 9500 Gilman Drive, UC San Diego, La Jolla, CA 92093, USA
| | - Fei Ning
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Chaoran Cheng
- Department of Computer Science, College of Computing Sciences, New Jersey Institute of Technology, Neswark, NJ 07102, USA
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | | | - Yan Sun
- Lanzhou Institute of Husbandry and Pharmaceutical Science of CAAS, Lanzhou 730050, China
| | - Yanyi Huang
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics (ICG), College of Chemistry, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Dong Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaoyu Hu
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Zhi Wei
- Department of Computer Science, College of Computing Sciences, New Jersey Institute of Technology, Neswark, NJ 07102, USA
| | - Wei Wang
- Department of Chemistry and Biochemistry, 9500 Gilman Drive, UC San Diego, La Jolla, CA 92093, USA
| | - Xuebin Liao
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Beijing Advanced Innovation Center for Human Brain Protection, Tsinghua University, Beijing 100084, China
| |
Collapse
|
322
|
Wang Z, Wu X. Study and analysis of antitumor resistance mechanism of PD1/PD-L1 immune checkpoint blocker. Cancer Med 2020; 9:8086-8121. [PMID: 32875727 PMCID: PMC7643687 DOI: 10.1002/cam4.3410] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 12/16/2022] Open
Abstract
Immunocheckpoint proteins of tumor infiltrating lymphocytes play an important role in tumor prognosis in the course of tumor clinicopathology. PD‐1 (Programmed cell death protein 1) is an important immunosuppressive molecule. By binding to PD‐L1 (programmed cell death‐ligand 1), it blocks TCR and its costimulus signal transduction, inhibits the activation and proliferation of T cells, depletes the function of effector T cells, and enables tumor cells to achieve immune escape. In recent years, immunocheckpoint blocking therapy targeting the PD‐1/PD‐L1 axis has achieved good results in a variety of malignant tumors, pushing tumor immunotherapy to a new milestone, such as anti‐PD‐1 monoclonal antibody Nivolumab, Pembrolizumab, and anti‐PD‐L1 monoclonal antibody Atezolizumab, which are considered as potential antitumor drugs. It was found in clinical use that some patients obtained long‐term efficacy, but most of them developed drug resistance recurrence in the later stage. The high incidence of drug resistance (including primary and acquired drug resistance) still cannot be ignored, which limited its clinical application and became a new problem in this field. Due to tumor heterogeneity, current limited research shows that PD‐1 or PD‐L1 monoclonal antibody drug resistance may be related to the following factors: mutation of tumor antigen and antigen presentation process, multiple immune checkpoint interactions, immune microenvironment changes dynamically, activation of oncogenic pathways, gene mutation and epigenetic changes of key proteins in tumors, tumor competitive metabolism, and accumulation of metabolites, etc, mechanisms of resistance are complex. Therefore, it is the most urgent task to further elucidate the mechanism of immune checkpoint inhibitor resistance, discover multitumor universal biomarkers, and develop new target agents to improve the response rate of immunotherapy in patients. In this study, the mechanism of anti‐PD‐1/PD‐L1 drug resistance in tumors, the potential biomarkers for predicting PD‐1 acquired resistance, and the recent development of combination therapy were reviewed one by one. It is believed that, based on the complex mechanism of drug resistance, it is of no clinical significance to simply search for and regulate drug resistance targets, and it may even produce drug resistance again soon. It is speculated that according to the possible tumor characteristics, three types of treatment methods should be combined to change the tumor microenvironment ecology and eliminate various heterogeneous tumor subsets, so as to reduce tumor drug resistance and improve long‐term clinical efficacy.
Collapse
Affiliation(s)
- Zhengyi Wang
- GCP Center of Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital Medical Sciences, Chengdu City, Sichuan Province, China.,Institute of Laboratory Animals of Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu City, Sichuan Province, China
| | - Xiaoying Wu
- Ministry of Education and Training, Second People's Hospital, Chengdu City, Sichuan Province, China
| |
Collapse
|
323
|
Hu ZY, Huang WY, Zhang L, Huang B, Chen SC, Li XL. Expression of AKT and p-AKT protein in lung adenocarcinoma and its correlation with PD-L1 protein and prognosis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1172. [PMID: 33241021 PMCID: PMC7576079 DOI: 10.21037/atm-20-5865] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background The PI3K/AKT/mTOR signaling pathway were significantly associated with EGFR mutation in lung adenocarcinoma (LUAD), but its correlation with PD-L1 protein and prognosis are not clear. The aim of this study was to evaluate the expression of AKT and phosphorylated AKT (p-AKT) in LUAD and its correlation with programmed death ligand-1 (PD-L1); and to analyze the factors affecting LUAD prognosis. Methods The expression of AKT, p-AKT, and PD-L1 was examined using immunohistochemistry in LUAD tissues from 110 patients who underwent surgical treatment. Results AKT protein expression was examined in 64.5% (71/110) of the LUAD samples, and p-AKT protein expression was examined in 44.5% (49/110) of the LUAD samples. The positive rate of PD-L1 at TC1/2/3 was 38.2% (42/110). AKT and p-AKT expression was significantly associated with epidermal growth factor receptor (EGFR) mutation (P=0.016, P=0.014 respectively). Pearson's correlation analysis indicated a negative correlation of p-AKT with PD-L1 protein (P=0.022). Out of the 62 patients with EGFR mutation, the expression of PD-L1 was negatively correlated with that of p-AKT protein (P=0.032). The expressions of AKT and p-AKT were not associated with prognosis. Multivariate analysis showed that tumor-node-metastasis (TNM) stage (P=0.013) and differentiation (P=0.046) were independent prognostic factors for overall survival. Conclusions PI3K/AKT/mTOR in the downstream pathway of EGFR may negatively regulate the expression of PD-L1, which may partly explain why patients with EGFR mutation respond poorly to PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Zhi-Ying Hu
- Department of Thoracic Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China.,Department of Respiratory and Critical Care Medicine, Dalian Third People's Hospital, Dalian, China
| | - Wan-Yi Huang
- Department of aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Lei Zhang
- Department of Oncology, Shenyang Fifth People Hospital, Shenyang, China
| | - Bo Huang
- Department of Pathology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Shu-Chen Chen
- Department of Thoracic Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Xiao-Ling Li
- Department of Thoracic Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
324
|
Shang X, Li J, Wang H, Li Z, Lin J, Chen D, Wang H. CMTM6 is positively correlated with PD-L1 expression and immune cells infiltration in lung squamous carcinoma. Int Immunopharmacol 2020; 88:106864. [PMID: 32866782 DOI: 10.1016/j.intimp.2020.106864] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/13/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The aim of this study was to clarify the association between CMTM6 and PD-L1 expression as well as microenvironment in lung squamous carcinoma (LUSC). MATERIAL AND METHODS Using Spearman's correlation and Tumor Immune Estimation Resource (TIMER), we analyzed the relationship between CMTM6 and PD-L1 mRNA in LUSC. Immunohistochemistry (IHC) assay was applied to validate the correlation between CMTM6 and PD-L1 protein level in 80 LUSC samples originated from Shandong Provincial Hospital. Then, using The Cancer Genome Atlas (TCGA) database and fisher test, we analyzed the differential mutation genes in high and low CMTM6 expression group. TISIDB was used to explore the distribution of CMTM6 across immune- and molecular-subtypes. TCGA database and Gene Set variation analysis (GSVA) were used to analyze the relationship between CMTM6 and immune genes, immune related pathways. RESULT Positive correlation between CMTM6 and PD-L1 in mRNA and protein level was found in LUSC patients. More gene mutations were found in CMTM6 high expression group compared with low expression group. Meanwhile, we also found the correlation between CMTM6 expression and molecular subtypes, immune genes, immune related pathways. Furthermore, our result revealed that B cells memory, T cells memory testing, T cells folicular helper, macrophages M0, macrophages M1 and neutrophils varied significantly between patients with CMTM6 high and low expression group. Finally, we found that CMTM6 expression was positively related to CD8 + T cell, macrophage, neutrophil and dendtritic cell (all, P < 0.05) and negatively related to CD4 + T cell (P = 0.018). CONCLUSION CMTM6 is positively associated with PD-L1 expression and correlates with infiltration of immune cells in microenvironment of lung squamous carcinoma.
Collapse
Affiliation(s)
- Xiaoling Shang
- Department of Clinical Laboratory, Shandong University, Jinan 250012, China; Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Jia Li
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai 200032, China
| | - Hui Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Zhenxiang Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Jiamao Lin
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Dawei Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China.
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China.
| |
Collapse
|
325
|
Nie H, Bu F, Xu J, Li T, Huang J. 29 immune-related genes pairs signature predict the prognosis of cervical cancer patients. Sci Rep 2020; 10:14152. [PMID: 32843657 PMCID: PMC7447790 DOI: 10.1038/s41598-020-70500-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/23/2020] [Indexed: 12/24/2022] Open
Abstract
To screen the key immune genes in the development of cervical cancer, construct immune related gene pairs (IRGPs), and evaluate their influence on the prognosis of cervical cancer. Tumor Genome Atlas (TCGA) database and geo database were downloaded as training set and validation set respectively, and immune related gene data were downloaded from immport. IRGPs model is established by machine learning, and the model is analyzed and evaluated. Using the Uclcan to analyze the immune genes expression in cervical cancer, and to further explore the association with the expression level and the clinical stage and prognosis of cervical cancer. According to the analysis of training set, we identified 29 IRGPs as key gene pairs and constructed the model. The AUC value of the model was greater than 0.9, and the model group survival rate was conspicuous different (P < 0.001). The reliability of the model was confirmed in the validation group. Our IRGPs play an important role in the occurrence and development of cervical cancer, and can be used as a prognostic marker and potential new target of cervical cancer.
Collapse
Affiliation(s)
- Han Nie
- Department of Vascular Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi Provence, China
| | - Fanqin Bu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi Provence, China
| | - Jiasheng Xu
- Department of Vascular Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi Provence, China
| | - Taoshen Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi Provence, China
| | - Jun Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi Provence, China.
| |
Collapse
|
326
|
Morand S, Stanbery L, Walter A, Rocconi RP, Nemunaitis J. BRCA1/2 Mutation Status Impact on Autophagy and Immune Response: Unheralded Target. JNCI Cancer Spectr 2020; 4:pkaa077. [PMID: 33409454 PMCID: PMC7771003 DOI: 10.1093/jncics/pkaa077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/27/2022] Open
Abstract
BRCA1 and possibly BRCA2 proteins may relate to the regulation of autophagy. Autophagy plays a key role in immune response from both a tumor and immune effector cell standpoint. In cells with BRCA mutations, increased autophagy leads to elevated expression of major histocompatibility complex class II but may cause subclonal neoantigen presentation, which may impair the immune response related to clonal neoantigen visibility. We review evidence of BRCA1/2 regulation of autophagy, immune response, and antigen presentation.
Collapse
Affiliation(s)
- Susan Morand
- Department of Internal Medicine, University of Toledo, Toledo, OH, USA
| | | | | | - Rodney P Rocconi
- University of South Alabama - Mitchell Cancer Institute, Mobile, AL, USA
| | | |
Collapse
|
327
|
Lotfinejad P, Kazemi T, Mokhtarzadeh A, Shanehbandi D, Jadidi Niaragh F, Safaei S, Asadi M, Baradaran B. PD-1/PD-L1 axis importance and tumor microenvironment immune cells. Life Sci 2020; 259:118297. [PMID: 32822718 DOI: 10.1016/j.lfs.2020.118297] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/10/2020] [Accepted: 08/15/2020] [Indexed: 12/23/2022]
Abstract
Triple-negative breast cancer (TNBC) is heterogeneous cancer with poor prognosis among the other breast tumors. Rapid recurrence and increased progression rate could be reasons for the poor prognosis of this type of breast cancer. Recently, because of the lack of specific targets in multiple cancer treatment, immune checkpoint blockade therapies with targeting PD-1/PD-L1 axis have displayed significant advances and improved survival. Among different types of breast cancers, TNBC is considered more immunogenic with high T-cell and other immune cells infiltration compared to other breast cancer subtypes. This immunogenic characteristic of TNBC is a beneficial marker in the immunotherapy of these tumors. Clinical studies with a focus on immune checkpoint therapy have demonstrated promising results in TNBC treatment. In this review, we summarize clinical trials with the immunotherapy-based treatment of different cancers and also discuss the interaction between infiltrating immune cells and breast tumor microenvironment. In addition, we focus on the signaling pathway that controls PD-L1 expression and continues with CAR T-cell therapy and siRNA as novel strategies and potential tools in targeted therapy.
Collapse
Affiliation(s)
- Parisa Lotfinejad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
328
|
Voli F, Valli E, Lerra L, Kimpton K, Saletta F, Giorgi FM, Mercatelli D, Rouaen JRC, Shen S, Murray JE, Ahmed-Cox A, Cirillo G, Mayoh C, Beavis PA, Haber M, Trapani JA, Kavallaris M, Vittorio O. Intratumoral Copper Modulates PD-L1 Expression and Influences Tumor Immune Evasion. Cancer Res 2020; 80:4129-4144. [PMID: 32816860 DOI: 10.1158/0008-5472.can-20-0471] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/19/2020] [Accepted: 08/03/2020] [Indexed: 11/16/2022]
Abstract
Therapeutic checkpoint antibodies blocking programmed death receptor 1/programmed death ligand 1 (PD-L1) signaling have radically improved clinical outcomes in cancer. However, the regulation of PD-L1 expression on tumor cells is still poorly understood. Here we show that intratumoral copper levels influence PD-L1 expression in cancer cells. Deep analysis of the The Cancer Genome Atlas database and tissue microarrays showed strong correlation between the major copper influx transporter copper transporter 1 (CTR-1) and PD-L1 expression across many cancers but not in corresponding normal tissues. Copper supplementation enhanced PD-L1 expression at mRNA and protein levels in cancer cells and RNA sequencing revealed that copper regulates key signaling pathways mediating PD-L1-driven cancer immune evasion. Conversely, copper chelators inhibited phosphorylation of STAT3 and EGFR and promoted ubiquitin-mediated degradation of PD-L1. Copper-chelating drugs also significantly increased the number of tumor-infiltrating CD8+ T and natural killer cells, slowed tumor growth, and improved mouse survival. Overall, this study reveals an important role for copper in regulating PD-L1 and suggests that anticancer immunotherapy might be enhanced by pharmacologically reducing intratumor copper levels. SIGNIFICANCE: These findings characterize the role of copper in modulating PD-L1 expression and contributing to cancer immune evasion, highlighting the potential for repurposing copper chelators as enhancers of antitumor immunity. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/19/4129/F1.large.jpg.
Collapse
Affiliation(s)
- Florida Voli
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Emanuele Valli
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Luigi Lerra
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia
| | - Kathleen Kimpton
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW Sydney, New South Wales, Sydney, Australia
| | - Federica Saletta
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Federico M Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Jourdin R C Rouaen
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia
| | - Sylvie Shen
- Cord & Marrow Transplant Facility, Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia
| | - Jayne E Murray
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia
| | - Aria Ahmed-Cox
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW Sydney, New South Wales, Sydney, Australia
| | - Giuseppe Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Chelsea Mayoh
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Paul A Beavis
- Rosie Lew Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia
| | - Joseph A Trapani
- Rosie Lew Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia. .,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW Sydney, New South Wales, Sydney, Australia
| | - Orazio Vittorio
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, New South Wales, Australia. .,School of Women's and Children's Health, UNSW Sydney, Sydney, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, UNSW Sydney, New South Wales, Sydney, Australia
| |
Collapse
|
329
|
Asai A, Yasuoka H, Matsui M, Tsuchimoto Y, Fukunishi S, Higuchi K. Programmed Death 1 Ligand Expression in the Monocytes of Patients with Hepatocellular Carcinoma Depends on Tumor Progression. Cancers (Basel) 2020; 12:E2286. [PMID: 32824016 PMCID: PMC7465257 DOI: 10.3390/cancers12082286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/05/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022] Open
Abstract
Monocytes (CD14+ cells) from advanced-stage hepatocellular carcinoma (HCC) patients express programmed death 1 ligand (PD-L)/PD-1 and suppress the host antitumor immune response. However, it is unclear whether cancer progression is associated with CD14+ cells. We compared CD14+ cell properties before and after cancer progression in the same HCC patients and examined their role in antitumor immunity. CD14+ cells were isolated from 15 naïve early-stage HCC patients before treatment initiation and after cancer progression to advanced stages. Although CD14+ cells from patients at early HCC stages exhibited antitumor activity in humanized murine chimera, CD14+ cells from the same patients after progression to advanced stages lacked this activity. Moreover, CD14+ cells from early HCC stages scantly expressed PD-L1 and PD-L2 and produced few cytokines, while CD14+ cells from advanced stages showed increased PD-L expression and produced IL-10 and CCL1. CD14+ cells were also isolated from five naïve advanced-stage HCC patients before treatment as well as after treatment-induced tumor regression. The CD14+ cells from patients with advanced-stage HCC expressed PD-L expressions, produced IL-10 and CCL1, and exhibited minimal tumoricidal activity. After treatment-induced tumor regression, CD14+ cells from the same patients did not express PD-Ls, failed to produce cytokines, and recovered tumoricidal activity. These results indicate that PD-L expression as well as CD14+ cell phenotype depend on the tumor stage in HCC patients. PD-L expressions of monocytes may be used as a new marker in the classification of cancer progression in HCC.
Collapse
Affiliation(s)
- Akira Asai
- The Second Department of Internal Medicine, Osaka Medical College, Takatsuki 5698686, Japan; (H.Y.); (M.M.); (Y.T.); (S.F.); (K.H.)
| | | | | | | | | | | |
Collapse
|
330
|
Tumor suppressive activity of miR-424-5p in breast cancer cells through targeting PD-L1 and modulating PTEN/PI3K/AKT/mTOR signaling pathway. Life Sci 2020; 259:118239. [PMID: 32784058 DOI: 10.1016/j.lfs.2020.118239] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022]
Abstract
AIMS MicroRNAs (miRs) are key modulators of cellular processes such as proliferation, apoptosis, as well as anti-cancer immune responses. Here, we evaluated the role of miR-424-5p in breast cancer (BC) and investigated its effects on T cell-related immune response. MAIN METHODS BC tissues and cell lines were prepared and the expression of miR-424-5p and PD-L1, as well as the underlying molecular pathways, were assessed via qRT-PCR and western blotting. The MTT assay and flow cytometry were used to assess the effect of miR-424-5p on proliferation, apoptosis, autophagy, and cell cycle progression. The co-culture of T cells with MDA-MB-231 was performed for evaluating the role of miR-424-5p in rescuing T cell exhaustion. KEY FINDINGS The results indicated the down-regulation of miR-424-5p and up-regulation of PD-L1 expression in BC tissue specimens. MiR-424-5p transfection into PD-L1 overexpressing MDA-MB-231 cells decreased the expression of PD-L1. Also, miR-424-5p could reduce MDA-MB-231 cell viability through modulating apoptosis and autophagy pathways. Furthermore, miR-424-5p transfection leads to decreased colony formation and increased cell number at the G2/M phase. Western blot analysis illustrated that miR-424-5p could exert its anti-proliferative effect via modulating PTEN/PI3K/AKT/mTOR pathway. Moreover, it was demonstrated that suppression of PD-L1 by miR-424-5p could participate in regulating the expression of effector cytokines in T cells. SIGNIFICANCE MiR-424-5p could be considered as a potential tumor-suppressor miR in regulating BC cellular growth, apoptosis, and T cell-related immune response through targeting PD-L1, and its downstream mediators. Therefore, we recognized miR-424-5p as a promising candidate for miR restoration therapy in BC patients.
Collapse
|
331
|
Yoon MS. Nanotechnology-Based Targeting of mTOR Signaling in Cancer. Int J Nanomedicine 2020; 15:5767-5781. [PMID: 32821100 PMCID: PMC7418174 DOI: 10.2147/ijn.s254574] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) is a master regulator of cell growth and metabolism, which is activated in response to intra- and extracellular signals, including nutrients, growth factors, and cellular energy levels. The frequent dysregulation of mTOR signaling in cancer makes it an attractive therapeutic target, and several types of mTOR inhibitors have been developed. Nanoparticle-based mTOR modulators are predicted to target various cancers and deliver as well as release drugs in a controlled manner, resulting in enhanced bioavailability and reduced side effects. This mini-review is focused on the molecular mechanism of nanoparticle-based mTOR modulator action as well as the current development of mTOR inhibitors using nanoparticles. Understanding the biological function of nanoparticle-based mTOR modulators will contribute to the development of efficient nano-therapeutics for the treatment of cancers.
Collapse
Affiliation(s)
- Mee-Sup Yoon
- Department of Molecular Medicine, School of Medicine, Lee Gil Ya Cancer and Diabetes Institute, Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
332
|
Mukherji R, Marshall JL, Seeber A. Genomic Alterations and Their Implications on Survival in Nonmetastatic Colorectal Cancer: Status Quo and Future Perspectives. Cancers (Basel) 2020; 12:E2001. [PMID: 32707813 PMCID: PMC7465976 DOI: 10.3390/cancers12082001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 02/08/2023] Open
Abstract
The selection of treatment according to genomic alterations is a standard approach in metastatic colorectal cancer but is only starting to have an impact in the earlier stages of the disease. The status if genes like KRAS, BRAF, and MMR has substantial survival implications, and concerted research efforts have revolutionized treatment towards precision oncology. In contrast, a genomic-based approach has not changed the adjuvant setting after curative tumor-resection in the daily routine so far. This review focuses on the current knowledge regarding prognostic and predictive genomic biomarkers in patients with locally advanced nonmetastasized colorectal cancer. Furthermore, we provide an outlook on future challenges for a personalized adjuvant treatment approach in patients with colorectal cancer.
Collapse
Affiliation(s)
- Reetu Mukherji
- Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - John L. Marshall
- Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Andreas Seeber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck A-6020, Austria;
| |
Collapse
|
333
|
Umemura S, Zhu J, Chahine JJ, Kallakury B, Chen V, Kim IK, Zhang YW, Goto K, He Y, Giaccone G. Downregulation of CYLD promotes IFN-γ mediated PD-L1 expression in thymic epithelial tumors. Lung Cancer 2020; 147:221-228. [PMID: 32738418 DOI: 10.1016/j.lungcan.2020.07.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/09/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Recent genomic studies suggest the biological significance of the cylindromatosis (CYLD) gene in thymic epithelial tumors (TETs). CYLD is a crucial regulator of immune response, and we previously reported that CYLD mutation is associated with high PD-L1 expression in thymic carcinoma. Therefore, we wanted to explore the role and mechanism of CYLD in regulating PD-L1 expression in TETs. MATERIALS AND METHODS The role of CYLD in PD-L1 expression was assessed by knockdown of CYLD in TET cells upon stimulation with interferon gamma (IFN-γ), tumor necrosis factor-α (TNF-α) or polyinosinic-polycytidylic acid (poly I:C). The molecular mechanism was investigated through analysis of downstream molecules in the STAT1/IRF1 pathway. Moreover, the clinical correlation between low CYLD and high PD-L1 expression, and the clinical impact of CYLD expression were evaluated in tissue microarrays of 105 TET cases. RESULTS CYLD knockdown significantly enhanced the expression of PD-L1 in presence of IFN-γ stimulation in most TET cell lines. However, this phenomenon was not observed in presence of TNF-α stimulation. CYLD knockdown upregulated IFN-γ mediated activation of the STAT1/IRF1 axis, which in turn induced PD-L1 expression. Interestingly, we found a significant association between low CYLD expression and ≥ 50 % PD-L1 expression (p = 0.001). In addition, the average proportion of tumor cells exhibiting PD-L1 staining was significantly higher in the low CYLD expression group (24.7 %) than in the high CYLD expression group (5.2 %) (p = 0.005). There was no correlation between CYLD expression and the frequency of pre-existing paraneoplastic auto-immune diseases. In advanced stages (III/IV), the low CYLD expressing group had numerically worse survival than the high CYLD group (log-rank p = 0.089). CONCLUSIONS Our findings provide insight into the mechanism of regulation of PD-L1 expression by CYLD in TET cells. Tumors with low CYLD expression could be potential targets for PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Shigeki Umemura
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA; Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Jianquan Zhu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA; Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Lung Cancer Center, Tianjin Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Joeffrey J Chahine
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA
| | - Bhaskar Kallakury
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA
| | - Vincent Chen
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA
| | - In-Kyu Kim
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA
| | - Yu-Wen Zhang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA; Department of Cell Biology, University of Virginia, VA, USA
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Yongfeng He
- Meyer Cancer Center, Weill Cornel Medicine, NY, USA
| | - Giuseppe Giaccone
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA; Meyer Cancer Center, Weill Cornel Medicine, NY, USA.
| |
Collapse
|
334
|
Pei XD, Yao HL, Shen LQ, Yang Y, Lu L, Xiao JS, Wang XY, He ZL, Jiang LH. α-Cyperone inhibits the proliferation of human cervical cancer HeLa cells via ROS-mediated PI3K/Akt/mTOR signaling pathway. Eur J Pharmacol 2020; 883:173355. [PMID: 32687921 DOI: 10.1016/j.ejphar.2020.173355] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 01/06/2023]
Abstract
Cervical cancer is the fourth leading killer of female cancer patients worldwide. Each year more than half a million women are diagnosed with cervical cancer and the disease results in over 300, 000 deaths. α-Cyperone is known as the principal active ingredient in the Cyperus rotundus (Family: Cyperaceae). However, the effects of α-Cyperone on cancers, especially on cervical cancer, are yet to be explored. In the present study, the underlying mechanism of the anti-tumor activity of α-Cyperone against HeLa cells was investigated. The results showed that α-Cyperone inhibited proliferation and induced apoptosis in HeLa cells. Mechanistically, α-Cyperone promoted HeLa cells apoptosis via a mitochondrial apoptotic pathway, which was proved by increased level of intracellular reactive oxygen species (ROS) and upregulated expression of cytochrome c, cleaved caspase-3, PARP, and Bax. Further RNA-sequencing revealed α-Cyperone inhibited the activation of PI3K/Akt/mTOR signaling pathway in HeLa cells, which confirmed by PI3K inhibitor and agonist. The PI3K inhibitor (LY294002) synergized with α-Cyperone in arresting the growth of HeLa cells, whereas the PI3K agonist (IGF-1) abrogated such an effect. Interestingly, the expression of PD-L1 was attenuated by both α-Cyperone and LY294002, while the supplement of IGF-1 rescued the low expression of PD-L1. In conclusion, our results reveal that the inhibitory effect of α-Cyperone on HeLa cell growth is triggered via the ROS-mediated PI3K/Akt/mTOR signaling pathway and closely related to a decline in the PD-L1 expression.
Collapse
Affiliation(s)
- Xiao-Dong Pei
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563100, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China; College of Light Industry and Food Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Hong-Liang Yao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Drug Synthesis and Evaluation Center, Guangdong Institute of Applied Biological Resources, Guangdong, PR China
| | - Li-Qun Shen
- Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Guangxi University for Nationalities, Nanning, 530006, PR China
| | - Yang Yang
- Department of Pharmacology, Guilin Medical University, Guilin, Guangxi, 541000, PR China
| | - Lan Lu
- Sichuan Industrial Institute of Antibiotics, Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Chengdu University, 610106, Chengdu, PR China
| | - Jun-Song Xiao
- Beijing Higher Institution Engineering Research Center of Food Additives and Ingredients, Beijing Technology and Business University-BTBU, Beijing, 100048, PR China
| | - Xin-Yu Wang
- College of Light Industry and Food Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Zhi-Long He
- College of Light Industry and Food Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Li-He Jiang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563100, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China; College of Light Industry and Food Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China; Medical College, Guangxi University, Nanning, 530004, PR China; School of Basic Medical Science, YouJiang Medical University for Nationaties, No. 98 Chengxiang Road, Baise, Guangxi, 533000, PR China.
| |
Collapse
|
335
|
Kongtawelert P, Wudtiwai B, Shwe TH, Pothacharoen P, Phitak T. Inhibition of programmed death ligand 1 (PD-L1) expression in breast cancer cells by sesamin. Int Immunopharmacol 2020; 86:106759. [PMID: 32663768 DOI: 10.1016/j.intimp.2020.106759] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/17/2020] [Accepted: 06/26/2020] [Indexed: 12/28/2022]
Abstract
Programmed death ligand 1 (PD-L1) is overexpressed in some metastatic breast cancer subtypes, specifically triple-negative breast cancer (TNBC). This feature can assist in the eradication of anti-tumor immunity, thereby enhancing the survival of the tumor. This study aims to explore how sesamin affects PD-L1 expression in breast cancer cells and its related molecular mechanisms. We found high levels of expression of PD-L1 in both mRNA and protein levels in the TNBC cell line, MDA-MB231, but not in the luminal type-breast cancer cell line, MCF-7. We then demonstrated the tumor suppressive effect of sesamin, which induced the inhibition of cell proliferation in MDA-MB231 cells. Additionally, sesamin triggered PD-L1 downregulation (both mRNA and protein) through the inhibition of AKT, NF-κB and JAK/Stat signaling in MDA-MB231 cells. Moreover, the migration ability of MDA-MB231 cells was effectively diminished by sesamin via inhibition of the activation of MMP-9 and MMP-2. In summary, this study demonstrated that sesamin suppresses MDA-MB231 breast cancer cells' proliferation and migration; and decreases the expression of PD-L1 via the downregulation of AKT, NF-κB, and JAK/Stat signaling. Therefore, sesamin may be an effective alternative and novel therapeutic option for immunotherapy in breast cancer cells with high PD-L1 expression.
Collapse
Affiliation(s)
- Prachya Kongtawelert
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Benjawan Wudtiwai
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thuzar Hla Shwe
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Peraphan Pothacharoen
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thanyaluck Phitak
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
336
|
Zheng NN, Zhou M, Sun F, Huai MX, Zhang Y, Qu CY, Shen F, Xu LM. Combining protein arginine methyltransferase inhibitor and anti-programmed death-ligand-1 inhibits pancreatic cancer progression. World J Gastroenterol 2020; 26:3737-3749. [PMID: 32774054 PMCID: PMC7383845 DOI: 10.3748/wjg.v26.i26.3737] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 06/02/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Immunotherapy targeting programmed death-1 (PD-1) or programmed death-ligand-1 (PD-L1) has been shown to be effective in a variety of malignancies but has poor efficacy in pancreatic ductal adenocarcinoma (PDAC). Studies have shown that PD-L1 expression in tumors is an important indicator of the efficacy of immunotherapy. Tumor cells usually evade chemotherapy and host immune surveillance by epigenetic changes. Protein arginine methylation is a common posttranslational modification. Protein arginine methyltransferase (PRMT) 1 is deregulated in a wide variety of cancer types, whose biological role in tumor immunity is undefined.
AIM To investigate the combined effects and underlying mechanisms of anti-PD-L1 and type I PRMT inhibitor in pancreatic cancer in vivo.
METHODS PT1001B is a novel type I PRMT inhibitor with strong activity and good selectivity. A mouse model of subcutaneous Panc02-derived tumors was used to evaluate drug efficacy, toxic and side effects, and tumor growth in vivo. By flow cytometry, we determined the expression of key immune checkpoint proteins, detected the apoptosis in tumor tissues, and analyzed the immune cells. Immunohistochemistry staining for cellular proliferation-associated nuclear protein Ki67, TUNEL assay, and PRMT1/PD-L1 immunofluorescence were used to elucidate the underlying molecular mechanism of the antitumor effect.
RESULTS Cultured Panc02 cells did not express PD-L1 in vitro, but tumor cells derived from Panc02 transplanted tumors expressed PD-L1. The therapeutic efficacy of anti-PD-L1 mAb was significantly enhanced by the addition of PT1001B as measured by tumor volume (1054.00 ± 61.37 mm3vs 555.80 ± 74.42 mm3, P < 0.01) and tumor weight (0.83 ± 0.06 g vs 0.38 ± 0.02 g, P < 0.05). PT1001B improved antitumor immunity by inhibiting PD-L1 expression on tumor cells (32.74% ± 5.89% vs 17.95% ± 1.92%, P < 0.05). The combination therapy upregulated tumor-infiltrating CD8+ T lymphocytes (23.75% ± 3.20% vs 73.34% ± 4.35%, P < 0.01) and decreased PD-1+ leukocytes (35.77% ± 3.30% vs 6.48% ± 1.08%, P < 0.001) in tumor tissue compared to the control. In addition, PT1001B amplified the inhibitory effect of anti-PD-L1 on tumor cell proliferation and enhanced the induction of tumor cell apoptosis. PRMT1 downregulation was correlated with PD-L1 downregulation.
CONCLUSION PT1001B enhances antitumor immunity and combining it with anti-PD-L1 checkpoint inhibitors provides a potential strategy to overcome anti-PD-L1 resistance in PDAC.
Collapse
Affiliation(s)
- Nan-Nan Zheng
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Min Zhou
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Fang Sun
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Man-Xiu Huai
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yi Zhang
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Chun-Ying Qu
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Feng Shen
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Lei-Ming Xu
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
337
|
Zhu H, Du C, Yuan M, Fu P, He Q, Yang B, Cao J. PD-1/PD-L1 counterattack alliance: multiple strategies for treating triple-negative breast cancer. Drug Discov Today 2020; 25:1762-1771. [PMID: 32663441 DOI: 10.1016/j.drudis.2020.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/25/2020] [Accepted: 07/06/2020] [Indexed: 01/01/2023]
Abstract
Despite extensive research into adjuvant and neoadjuvant chemotherapy, triple-negative breast cancer (TNBC) remains a common breast cancer (BC) subtype with poor prognosis. Given that it has higher immune cell infiltration, theoretically, it should be a protagonist of potential BC immunotherapies. However, only mild responses have been observed in monotherapy with anti-programmed death receptor-1/programmed death ligand-1 (PD-1/PD-L1) antibodies. In this review, we reappraise PD-1/PD-L1 inhibitor combination immunotherapy and effective experimental compounds, focusing the level of PD-L1 expression, neoantigens, abnormal signaling pathways, and tumor microenvironment signatures, to provide guidance for future clinical trials based on the molecular mechanisms involved.
Collapse
Affiliation(s)
- Haiying Zhu
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chengyong Du
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Meng Yuan
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Peifen Fu
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ji Cao
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
338
|
Tumor Cell-Intrinsic Immunometabolism and Precision Nutrition in Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12071757. [PMID: 32630618 PMCID: PMC7409312 DOI: 10.3390/cancers12071757] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
One of the greatest challenges in the cancer immunotherapy field is the need to biologically rationalize and broaden the clinical utility of immune checkpoint inhibitors (ICIs). The balance between metabolism and immune response has critical implications for overcoming the major weaknesses of ICIs, including their lack of universality and durability. The last decade has seen tremendous advances in understanding how the immune system's ability to kill tumor cells requires the conspicuous metabolic specialization of T-cells. We have learned that cancer cell-associated metabolic activities trigger shifts in the abundance of some metabolites with immunosuppressory roles in the tumor microenvironment. Yet very little is known about the tumor cell-intrinsic metabolic traits that control the immune checkpoint contexture in cancer cells. Likewise, we lack a comprehensive understanding of how systemic metabolic perturbations in response to dietary interventions can reprogram the immune checkpoint landscape of tumor cells. We here review state-of-the-art molecular- and functional-level interrogation approaches to uncover how cell-autonomous metabolic traits and diet-mediated changes in nutrient availability and utilization might delineate new cancer cell-intrinsic metabolic dependencies of tumor immunogenicity. We propose that clinical monitoring and in-depth molecular evaluation of the cancer cell-intrinsic metabolic traits involved in primary, adaptive, and acquired resistance to cancer immunotherapy can provide the basis for improvements in therapeutic responses to ICIs. Overall, these approaches might guide the use of metabolic therapeutics and dietary approaches as novel strategies to broaden the spectrum of cancer patients and indications that can be effectively treated with ICI-based cancer immunotherapy.
Collapse
|
339
|
Rennier K, Shin WJ, Krug E, Virdi G, Pachynski RK. Chemerin Reactivates PTEN and Suppresses PD-L1 in Tumor Cells via Modulation of a Novel CMKLR1-mediated Signaling Cascade. Clin Cancer Res 2020; 26:5019-5035. [PMID: 32605911 DOI: 10.1158/1078-0432.ccr-19-4245] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 05/18/2020] [Accepted: 06/26/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Chemerin (retinoic acid receptor responder 2, RARRES2) is an endogenous leukocyte chemoattractant that recruits innate immune cells through its receptor, ChemR23. RARRES2 is widely expressed in nonhematopoietic tissues and often downregulated across multiple tumor types compared with normal tissue. Recent studies show that augmenting chemerin in the tumor microenvironment significantly suppresses tumor growth, in part, by immune effector cells recruitment. However, as tumor cells express functional chemokine/chemoattractant receptors that impact their phenotype, we hypothesized that chemerin may have additional, tumor-intrinsic effects. EXPERIMENTAL DESIGN We investigated the effect of exogenous chemerin on human prostate and sarcoma tumor lines. Key signaling pathway components were elucidated using qPCR, Western blotting, siRNA knockdown, and specific inhibitors. Functional consequences of chemerin treatment were evaluated using in vitro and in vivo studies. RESULTS We show for the first time that human tumors exposed to exogenous chemerin significantly upregulate PTEN expression/activity, and concomitantly suppress programmed death ligand-1 (PD-L1) expression. CMKLR1 knockdown abrogated chemerin-induced PTEN and PD-L1 modulation, exposing a novel CMKLR1/PTEN/PD-L1 signaling cascade. Targeted inhibitors suggested signaling was occurring through the PI3K/AKT/mTOR pathway. Chemerin treatment significantly reduced tumor migration, while significantly increasing T-cell-mediated cytotoxicity. Chemerin treatment was as effective as both PD-L1 knockdown and the anti-PD-L1 antibody, atezolizumab, in augmenting T-cell-mediated tumor lysis. Forced expression of chemerin in human DU145 tumors significantly suppressed in vivo tumor growth, and significantly increased PTEN and decreased PD-L1 expression. CONCLUSIONS Collectively, our data show a novel link between chemerin, PTEN, and PD-L1 in human tumor lines, which may have a role in improving T-cell-mediated immunotherapies.
Collapse
Affiliation(s)
- Keith Rennier
- Division of Oncology, John T. Milliken Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Woo Jae Shin
- Division of Oncology, John T. Milliken Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ethan Krug
- Division of Oncology, John T. Milliken Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Gurpal Virdi
- Division of Oncology, John T. Milliken Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Russell K Pachynski
- Division of Oncology, John T. Milliken Department of Medicine, Washington University School of Medicine, St. Louis, Missouri. .,Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri.,The Bursky Center for Human Immunology & Immunotherapy Programs (CHiiPs), Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
340
|
Comprehensive Investigation into the Role of Ubiquitin-Conjugating Enzyme E2S in Melanoma Development. J Invest Dermatol 2020; 141:374-384. [PMID: 32603752 DOI: 10.1016/j.jid.2020.05.113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/27/2020] [Accepted: 05/03/2020] [Indexed: 12/21/2022]
Abstract
Ubiquitin-conjugating enzyme E2S (UBE2S) is involved in protein degradation and signal transduction, but its function in the development of melanoma is unclear. We focused on the role of UBE2S in melanoma development both in vitro and in vivo. UBE2S was overexpressed in malignant melanoma cells and tissues, and UBE2S expression was significantly different between tumor node metastasis staging T4 and T1/T2/T3. We designed UBE2S short hairpin RNA (shUBE2S) and transfected it into A375, SK-MEL-28, and MUM-2B cells using lentivirus. By whole-genome filtering, 247 genes and 265 genes were upregulated and downregulated, respectively, in shUBE2S-treated melanoma; these genes were mainly involved in immune reactions, apoptosis, DNA damage repair, and cell movement. The proliferation of melanoma cells was inhibited, apoptosis was increased, and cell cycle was arrested in G1/S in shUBE2S-treated melanoma. Expression of epithelial to mesenchymal transition-related proteins was significantly suppressed, and tumor growth was also suppressed in shUBE2S BALB/C nude mice. shUBE2S treatment may cause cell cycle arrest in G1/S phase, inhibit proliferation, induce apoptosis, and suppress tumor growth through DNA damage repair, epithelial to mesenchymal transition inhibition, protein kinase B-mTOR pathway, NF-κB signaling, and immune reactions, which provides a comprehensive understanding of the role of UBE2S in melanoma development and the need for advanced clinical research into UBE2S.
Collapse
|
341
|
Hao C, Chen G, Zhao H, Li Y, Chen J, Zhang H, Li S, Zhao Y, Chen F, Li W, Jiang WG. PD-L1 Expression in Glioblastoma, the Clinical and Prognostic Significance: A Systematic Literature Review and Meta-Analysis. Front Oncol 2020; 10:1015. [PMID: 32670884 PMCID: PMC7326811 DOI: 10.3389/fonc.2020.01015] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 05/22/2020] [Indexed: 01/11/2023] Open
Abstract
Background: The clinical and prognostic value of programmed death-ligand 1, PD-L1, in glioblastoma remains controversial. The present study aimed to identify the expression of PD-L1 for its prognostic value in glioblastoma. Methods: A comprehensive literature search was performed using the PubMed and CNKI databases. The overall survival (OS) and disease-free survival (DFS) of GBM was analyzed based on Hazard ratios (HRs) and 95% confidence intervals (CIs). Furthermore, Odds ratios (ORs) and 95% CIs were summarized for clinicopathological parameters. The statistical analysis was using RevMan 5.3 software. Results: The meta-analysis was performed by using total nine studies including 806 patients who had glioblastoma. The pooled results indicated that PD-L1 expression in tumor tissues was significantly related to a poor OS (HR = 1.63, 95%CI: 1.19–2.24, P = 0.003, random effects model) with heterogeneity (I2 = 51%). In subgroup analyses, PD-L1 positivity was significantly associated with a worse OS for patients of American and Asian regions, but not for those of European regions. Moreover, PD-L1 expression implied a trend toward the mutation status of the IDH1 gene [coding the Isocitrate Dehydrogenase (NADP(+))-1 protein] (HR = 9.92, 95%CI: 1.85–53.08, P = 0.007, fixed effects model). However, the prediction overall survival (OS) of the patients showed that PD-L1 expression was independent from other clinicopathological features, such as gender and age. Conclusions: Our analyses indicated that high expression of PD-L1 in glioblastoma tumor tissues is associated with poor survival of patients, and PD-L1 may act as a prognostic predictor and an effective therapeutic target for glioblastoma.
Collapse
Affiliation(s)
- Chengcheng Hao
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Gang Chen
- Beijing Qinglian Biotech, Co., Ltd., Beijing, China
| | - Huishan Zhao
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yan Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jianxin Chen
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hongmei Zhang
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shan Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yuze Zhao
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Feng Chen
- Department of Neuro-Oncology, Neurosurgery Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Neuro-Oncology, Neurosurgery Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, United Kingdom
| |
Collapse
|
342
|
Brassart-Pasco S, Dalstein V, Brassart B, Dewolf M, Clavel C, Oudart JB. Immunotherapy in non-small-cell lung cancer: from targeted molecules to resistance patterns. Pharmacogenomics 2020; 21:705-720. [PMID: 32567537 DOI: 10.2217/pgs-2020-0021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Immunotherapies are now considered as a pillar of non-small-cell lung cancer treatment. The main targets of immune-checkpoint inhibitors (ICI) are programmed cell death 1/programmed cell death ligand 1 and cytotoxic T-lymphocyte antigen 4, aiming at restoring antitumor immunity. Despite durable responses observed in some patients, all patients do not benefit from the treatment and almost all responders ultimately relapse after some time. In this review, we discuss the biomarkers that could be used to predict response to ICI, the current indications of ICI in non-small-cell lung cancer, the mechanisms inducing tumor-cell intrinsic or extrinsic resistance to ICI and finally, the potential treatment response monitoring.
Collapse
Affiliation(s)
- Sylvie Brassart-Pasco
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, 51100 Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire - MEDyC, 51100 Reims, France
| | - Véronique Dalstein
- Université de Reims Champagne Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, 51100 Reims, France.,CHU Reims, Service de Pathologie, 51100 Reims, France
| | - Bertrand Brassart
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, 51100 Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire - MEDyC, 51100 Reims, France
| | - Maxime Dewolf
- CHU Reims, Service des maladies respiratoires et allergiques, 51100 Reims, France
| | - Christine Clavel
- Université de Reims Champagne Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, 51100 Reims, France.,CHU Reims, Service de Pathologie, 51100 Reims, France
| | - Jean-Baptiste Oudart
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, 51100 Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire - MEDyC, 51100 Reims, France.,CHU Reims, Service de Biochimie-Pharmacologie-Toxicologie, 51100 Reims, France
| |
Collapse
|
343
|
Immuno-Metabolism and Microenvironment in Cancer: Key Players for Immunotherapy. Int J Mol Sci 2020; 21:ijms21124414. [PMID: 32575899 PMCID: PMC7352562 DOI: 10.3390/ijms21124414] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/15/2020] [Accepted: 06/19/2020] [Indexed: 12/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have changed therapeutic algorithms in several malignancies, although intrinsic and secondary resistance is still an issue. In this context, the dysregulation of immuno-metabolism plays a leading role both in the tumor microenvironment (TME) and at the host level. In this review, we summarize the most important immune-metabolic factors and how they could be exploited therapeutically. At the cellular level, an increased concentration of extracellular adenosine as well as the depletion of tryptophan and uncontrolled activation of the PI3K/AKT pathway induces an immune-tolerant TME, reducing the response to ICIs. Moreover, aberrant angiogenesis induces a hypoxic environment by recruiting VEGF, Treg cells and immune-suppressive tumor associated macrophages (TAMs). On the other hand, factors such as gender and body mass index seem to affect the response to ICIs, while the microbiome composition (and its alterations) modulates both the response and the development of immune-related adverse events. Exploiting these complex mechanisms is the next goal in immunotherapy. The most successful strategy to date has been the combination of antiangiogenic drugs and ICIs, which prolonged the survival of patients with non-small-cell lung cancer (NSCLC) and hepatocellular carcinoma (HCC), while results from tryptophan pathway inhibition studies are inconclusive. New exciting strategies include targeting the adenosine pathway, TAMs and the microbiota with fecal microbiome transplantation.
Collapse
|
344
|
Choi H, Deng J, Li S, Silk T, Dong L, Brea EJ, Houghton S, Redmond D, Zhong H, Boiarsky J, Akbay EA, Smith PD, Merghoub T, Wong KK, Wolchok JD. Pulsatile MEK Inhibition Improves Anti-tumor Immunity and T Cell Function in Murine Kras Mutant Lung Cancer. Cell Rep 2020; 27:806-819.e5. [PMID: 30995478 PMCID: PMC6719696 DOI: 10.1016/j.celrep.2019.03.066] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 02/01/2019] [Accepted: 03/18/2019] [Indexed: 01/01/2023] Open
Abstract
KRAS is one of the driver oncogenes in non-small-cell lung cancer (NSCLC) but remains refractory to current modalities of targeted pathway inhibition, which include inhibiting downstream kinase MEK to circumvent KRAS activation. Here, we show that pulsatile, rather than continuous, treatment with MEK inhibitors (MEKis) maintains T cell activation and enables their proliferation. Two MEKis, selumetinib and trametinib, induce T cell activation with increased CTLA-4 expression and, to a lesser extent, PD-1 expression on T cells in vivo after cyclical pulsatile MEKi treatment. In addition, the pulsatile dosing schedule alone shows superior anti-tumor effects and delays the emergence of drug resistance. Furthermore, pulsatile MEKi treatment combined with CTLA-4 blockade prolongs survival in mice bearing tumors with mutant Kras. Our results set the foundation and show the importance of a combinatorial therapeutic strategy using pulsatile targeted therapy together with immunotherapy to optimally enhance tumor delay and promote long-term anti-tumor immunity.
Collapse
Affiliation(s)
- Hyejin Choi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jiehui Deng
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - Shuai Li
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - Tarik Silk
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lauren Dong
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elliott J Brea
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA
| | - Sean Houghton
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David Redmond
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hong Zhong
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jonathan Boiarsky
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Esra A Akbay
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
| | - Paul D Smith
- Bioscience, iMed Oncology, AstraZeneca, CRUK Cambridge Institute, Cambridge CB2 0RE, UK
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA.
| | - Kwok-Kin Wong
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA.
| | - Jedd D Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
345
|
Roles for receptor tyrosine kinases in tumor progression and implications for cancer treatment. Adv Cancer Res 2020; 147:1-57. [PMID: 32593398 DOI: 10.1016/bs.acr.2020.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Growth factors and their receptor tyrosine kinases (RTKs), a group of transmembrane molecules harboring cytoplasm-facing tyrosine-specific kinase functions, play essential roles in migration of multipotent cell populations and rapid proliferation of stem cells' descendants, transit amplifying cells, during embryogenesis and tissue repair. These intrinsic functions are aberrantly harnessed when cancer cells undergo intertwined phases of cell migration and proliferation during cancer progression. For example, by means of clonal expansion growth factors fixate the rarely occurring driver mutations, which initiate tumors. Likewise, autocrine and stromal growth factors propel angiogenesis and penetration into the newly sprouted vessels, which enable seeding micro-metastases at distant organs. We review genetic and other mechanisms that preempt ligand-mediated activation of RTKs, thereby supporting sustained cancer progression. The widespread occurrence of aberrant RTKs and downstream signaling pathways in cancer, identifies molecular targets suitable for pharmacological intervention. We list all clinically approved cancer drugs that specifically intercept oncogenic RTKs. These are mainly tyrosine kinase inhibitors and monoclonal antibodies, which can inhibit cancer but inevitably become progressively less effective due to adaptive rewiring processes or emergence of new mutations, processes we overview. Similarly important are patient treatments making use of radiation, chemotherapeutic agents and immune checkpoint inhibitors. The many interfaces linking RTK-targeted therapies and these systemic or local regimens are described in details because of the great promise offered by combining pharmacological modalities.
Collapse
|
346
|
Chen L, Liu S, Tao Y. Regulating tumor suppressor genes: post-translational modifications. Signal Transduct Target Ther 2020; 5:90. [PMID: 32532965 PMCID: PMC7293209 DOI: 10.1038/s41392-020-0196-9] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 05/19/2020] [Accepted: 05/24/2020] [Indexed: 01/10/2023] Open
Abstract
Tumor suppressor genes cooperate with each other in tumors. Three important tumor suppressor proteins, retinoblastoma (Rb), p53, phosphatase, and tensin homolog deleted on chromosome ten (PTEN) are functionally associated and they regulated by post-translational modification (PTMs) as well. PTMs include phosphorylation, SUMOylation, acetylation, and other novel modifications becoming growing appreciated. Because most of PTMs are reversible, normal cells use them as a switch to control the state of cells being the resting or proliferating, and PTMs also involve in cell survival and cell cycle, which may lead to abnormal proliferation and tumorigenesis. Although a lot of studies focus on the importance of each kind of PTM, further discoveries shows that tumor suppressor genes (TSGs) form a complex "network" by the interaction of modification. Recently, there are several promising strategies for TSGs for they change more frequently than carcinogenic genes in cancers. We here review the necessity, characteristics, and mechanisms of each kind of post-translational modification on Rb, p53, PTEN, and its influence on the precise and selective function. We also discuss the current antitumoral therapies of Rb, p53 and PTEN as predictive, prognostic, and therapeutic target in cancer.
Collapse
Affiliation(s)
- Ling Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute, Central South University, 410078, Changsha, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China.
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute, Central South University, 410078, Changsha, Hunan, China.
- Hunan Key Laboratory of Early Diagnosis and Precision Therapy, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011, Changsha, China.
| |
Collapse
|
347
|
Wang F, Yu T, Ma C, Yuan H, Zhang H, Zhang Z. Prognostic Value of Programmed Cell Death 1 Ligand-1 in Patients With Bone and Soft Tissue Sarcomas: A Systemic and Comprehensive Meta-Analysis Based on 3,680 Patients. Front Oncol 2020; 10:749. [PMID: 32582532 PMCID: PMC7280448 DOI: 10.3389/fonc.2020.00749] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/20/2020] [Indexed: 12/27/2022] Open
Abstract
Background: Programmed cell death 1 ligand-1 (PD-L1) is an immune checkpoint molecule that acts to protect cancer cells from immune surveillance and is considered as a prognostic biomarker in several cancers, but the prognostic value of PD-L1 in bone and soft tissue sarcomas remains inconclusive. In the present meta-analysis, the clinicopathological and prognostic value of PD-L1 in sarcomas was evaluated. Method: We performed a systemic and comprehensive meta-analysis by searching the PubMed, Medline, Cochrane Library, EMBASE, and Web of Science databases up to October 31, 2019. Eligible articles were incorporated, and pooled hazard ratios (HRs) and odds ratios (ORs) with their 95% confidence intervals (CIs) were used to estimate the outcomes. Results: Thirty-six articles containing 39 independent studies with 3,680 bone and soft tissue sarcoma patients were included in our meta-analysis. The pooled results showed that PD-L1 overexpression could predict poor overall survival (HR 1.45, 95% CI 1.11–1.90, P < 0.01), metastasis-free survival (HR 1.58, 95% CI 1.14–2.19, P < 0.01), and event-free survival (HR 2.82, 95% CI 1.69–4.71, P < 0.01) in sarcomas. Furthermore, PD-L1 overexpression was correlated with a higher rate of tumor metastasis (OR 2.95, 95% CI 1.32–6.60, P < 0.01), a more advanced tumor grade (OR 3.63, 95% CI 2.55–5.16, P < 0.01), and more T lymphocyte infiltration (OR 5.55, 95% CI 2.86–10.76, P < 0.01). No obvious publication bias was observed, and the sensitivity analysis showed that our results were robust. Conclusion: The results of our meta-analysis indicate that high PD-L1 expression might serve as a valuable and predictive biomarker for adverse clinicopathological features and poor prognosis in patients with sarcoma.
Collapse
Affiliation(s)
- Feng Wang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Tao Yu
- Center for Translational Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Chengbin Ma
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Hongmou Yuan
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Haifei Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
348
|
Vidotto T, Melo CM, Castelli E, Koti M, Dos Reis RB, Squire JA. Emerging role of PTEN loss in evasion of the immune response to tumours. Br J Cancer 2020; 122:1732-1743. [PMID: 32327707 PMCID: PMC7283470 DOI: 10.1038/s41416-020-0834-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 02/10/2020] [Accepted: 03/18/2020] [Indexed: 12/31/2022] Open
Abstract
Mutations in PTEN activate the phosphoinositide 3-kinase (PI3K) signalling network, leading to many of the characteristic phenotypic changes of cancer. However, the primary effects of this gene on oncogenesis through control of the PI3K-AKT-mammalian target of rapamycin (mTOR) pathway might not be the only avenue by which PTEN affects tumour progression. PTEN has been shown to regulate the antiviral interferon network and thus alter how cancer cells communicate with and are targeted by immune cells. An active, T cell-infiltrated microenvironment is critical for immunotherapy success, which is also influenced by mutations in DNA damage repair pathways and the overall mutational burden of the tumour. As PTEN has a role in the maintenance of genomic integrity, it is likely that a loss of PTEN affects the immune response at two different levels and might therefore be instrumental in mediating failed responses to immunotherapy. In this review, we summarise findings that demonstrate how the loss of PTEN function elicits specific changes in the immune response in several types of cancer. We also discuss ongoing clinical trials that illustrate the potential utility of PTEN as a predictive biomarker for immune checkpoint blockade therapies.
Collapse
Affiliation(s)
- Thiago Vidotto
- Department of Genetics, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Camila Morais Melo
- Department of Genetics, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Erick Castelli
- Department of Pathology, Medicine School of Botucatu, Paulista State University, Botucatu, Brazil
| | - Madhuri Koti
- Cancer Biology and Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | | | - Jeremy A Squire
- Department of Genetics, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
349
|
Vinod N, Hwang D, Azam SH, Van Swearingen AED, Wayne E, Fussell SC, Sokolsky-Papkov M, Pecot CV, Kabanov AV. High-capacity poly(2-oxazoline) formulation of TLR 7/8 agonist extends survival in a chemo-insensitive, metastatic model of lung adenocarcinoma. SCIENCE ADVANCES 2020; 6:eaba5542. [PMID: 32596460 PMCID: PMC7299629 DOI: 10.1126/sciadv.aba5542] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/06/2020] [Indexed: 05/03/2023]
Abstract
About 40% of patients with non-small cell lung cancer (NSCLC) have stage IV cancer at the time of diagnosis. The only viable treatment options for metastatic disease are systemic chemotherapy and immunotherapy. Nonetheless, chemoresistance remains a major cause of chemotherapy failure. New immunotherapeutic modalities such as anti-PD-1 immune checkpoint blockade have shown promise; however, response to such strategies is highly variable across patients. Here, we show that our unique poly(2-oxazoline)-based nanomicellar formulation (PM) of Resiquimod, an imidazoquinoline Toll-like receptor (TLR) 7/8 agonist, had a superior tumor inhibitory effect in a metastatic model of lung adenocarcinoma, relative to anti-PD-1 therapy or platinum-based chemotherapy. Investigation of the in vivo immune status following Resiquimod PM treatment showed that Resiquimod-based stimulation of antigen-presenting cells in the tumor microenvironment resulted in the mobilization of an antitumor CD8+ immune response. Our study demonstrates the promise of poly(2-oxazoline)-formulated Resiquimod for treating metastatic NSCLC.
Collapse
Affiliation(s)
- Natasha Vinod
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
- Joint UNC/NC State Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599-7575, USA
| | - Duhyeong Hwang
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Salma H. Azam
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Amanda E. D. Van Swearingen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elizabeth Wayne
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Sloane Christian Fussell
- Department of Biology, Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marina Sokolsky-Papkov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Chad V. Pecot
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alexander V. Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
- Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow 119992, Russia
| |
Collapse
|
350
|
Jin J, Zhao Q. Emerging role of mTOR in tumor immune contexture: Impact on chemokine-related immune cells migration. Theranostics 2020; 10:6231-6244. [PMID: 32483450 PMCID: PMC7255024 DOI: 10.7150/thno.45219] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/17/2020] [Indexed: 12/27/2022] Open
Abstract
During the last few decades, cell-based anti-tumor immunotherapy emerged and it has provided us with a large amount of knowledge. Upon chemokines recognition, immune cells undergo rapid trafficking and activation in disease milieu, with immune cells chemotaxis being accompanied by activation of diverse intercellular signal transduction pathways. The outcome of chemokines-mediated immune cells chemotaxis interacts with the cue of mammalian target of rapamycin (mTOR) in the tumor microenvironment (TME). Indeed, the mTOR cascade in immune cells involves migration and infiltration. In this review, we summarize the available mTOR-related chemokines, as well as the characterized upstream regulators and downstream targets in immune cells chemotaxis and assign potential underlying mechanisms in each evaluated chemokine. Specifically, we focus on the involvement of mTOR in chemokine-mediated immune related cells in the balance between tumor immunity and malignancy.
Collapse
Affiliation(s)
- Jing Jin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Qijie Zhao
- Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, 646000, Sichuan, PR China
- Department of Pathophysiology, College of Basic Medical Science, Southwest Medical University, Luzhou, 646000, Sichuan, PR China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, Sichuan, PR China
| |
Collapse
|