1
|
Li A, Luo M, Liu X, Wu H, Liu X, Zhang Z, Zhang X. Toll-like receptor 3 activation enhances antitumor immune response in lung adenocarcinoma through NF-κB signaling pathway. Front Immunol 2025; 16:1585747. [PMID: 40406122 PMCID: PMC12095255 DOI: 10.3389/fimmu.2025.1585747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 04/21/2025] [Indexed: 05/26/2025] Open
Abstract
Background Toll-like receptor 3 (TLR3) is a pattern recognition receptor known to play a crucial role in the immune response to cancer. However, its effect on the efficacy of immunotherapy in lung adenocarcinoma (LUAD) remains unclear. This study aims to investigate the role of TLR3 in LUAD by examining its expression levels, prognostic significance, and impact on immune signaling pathways. Methods We analyzed the impact of TLR3 expression on the prognosis of lung adenocarcinoma patients using data from the Cancer Genome Atlas (TCGA) database and four additional cohorts (GSE72094, GSE30219, GSE50081 and GSE31210). Functional enrichment analyses were performed to compare molecular features between low and high TLR3 expression groups using gene set variation analysis (GSVA). We also examined the correlation between TLR3 and tumor mutation burden (TMB), immune infiltration, and PD-L1 expression. Further experimental validation was conducted using co-culture systems of LUAD cells and peripheral blood mononuclear cells (PBMCs) with PD1 inhibitors, and Western blot analysis to investigate the involvement of NF-κB signaling. Results TLR3 expression was significantly lower in LUAD tissues compared to normal tissues, with high TLR3 expression correlating with better survival outcomes across multiple cohorts. High TLR3 expression was associated with increased TMB and enhanced immune activation. Patients with high TLR3 expression exhibited higher immune checkpoint expression and immune cell infiltration. Experimental results showed that TLR3 agonists increased the susceptibility of LUAD cells to activated PBMCs under PD1 inhibitor therapy, inhibiting cell proliferation, migration, and invasion. Additionally, TLR3 has a strong positive correlation with MHC molecules and upregulated PD-L1 expression. NF-κB was identified as a key regulator of PD-L1 expression, with TLR3 agonists enhancing NF-κB and PD-L1 activity. Conclusion TLR3 enhances the anti-tumor immune response in LUAD by modulating NF-κB signaling and PD-L1 expression, making it a promising prognostic biomarker and therapeutic target. This study highlights the potential of TLR3 to improve immunotherapy outcomes, providing a comprehensive analysis of its role in LUAD and paving the way for novel therapeutic strategies targeting TLR3-mediated pathways.
Collapse
Affiliation(s)
- Ang Li
- School of Public Health, North China University of Science and Technology, Tangshan, China
- College of Life Science, North China University of Science and Technology, Tangshan, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, North China University of Science and Technology, Tangshan, China
| | - Man Luo
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Xiyao Liu
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Hongjiao Wu
- School of Public Health, North China University of Science and Technology, Tangshan, China
- College of Life Science, North China University of Science and Technology, Tangshan, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, North China University of Science and Technology, Tangshan, China
| | - Xiaoguang Liu
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Zhi Zhang
- Affliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
| | - Xuemei Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, China
- College of Life Science, North China University of Science and Technology, Tangshan, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
2
|
Tandyk A, Christ T. Analysis of pembrolizumab in microsatellite instability high (MSI-H) and deficient mismatch repair (dMMR) non-colorectal patients with metastatic cancer. J Oncol Pharm Pract 2025:10781552251331958. [PMID: 40183381 DOI: 10.1177/10781552251331958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Pembrolizumab was approved for the treatment of MSI-H/dMMR solid malignancies in a tumor agnostic approval in 2017. Treatment records for patients harboring MSI-H/dMMR status at our institution treated with pembrolizumab were reviewed for outcomes of duration of treatment, progression free survival, and overall survival. Adverse events and immunotherapy biomarkers of PDL-1 status and tumor mutation burden (TMB) were also assessed. Thirty patients were included: 13 with endometrial cancer, 5 non-colorectal gastrointestinal cancer, 2 ovarian or uterine cancer, 2 breast cancer, and 8 with other malignancies. The median duration of treatment was 11 months (range 0-35). Progression free survival and overall survival were not reached. Five patients (16.7%) experienced immune related adverse events, resulting in discontinuation of pembrolizumab in 3. PDL-1 expression and TMB did not appear to correspond to treatment outcomes. This analysis provides a real-world dataset of MSI-H/dMMR solid tumor patients being treated with pembrolizumab, outcomes of which are consistent with registry trials.
Collapse
Affiliation(s)
- Alexis Tandyk
- Rush MD Anderson Cancer Center Pharmacy, Chicago, USA
- UCHealth University of Colorado Hospital, Department of Pharmacy, Aurora, USA
| | - Trevor Christ
- Rush MD Anderson Cancer Center Pharmacy, Chicago, USA
| |
Collapse
|
3
|
Zhu Y, Zhu Y, Chen S, Cai Q. Identifying the cancer-associated fibroblast signature to predict the prognosis and immunotherapy response in patients with lung squamous cell carcinoma. Comput Methods Biomech Biomed Engin 2025; 28:326-336. [PMID: 38015040 DOI: 10.1080/10255842.2023.2287418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are an important component of the tumor microenvironment that contribute toward the development of tumors. This study aimed to establish a new algorithm based on CAF scores to predict the prognosis and immunotherapy response in patients with lung squamous cell carcinoma (LUSC). The RNA-seq data of LUSC patients were obtained from two databases and merged after removing inter-batch differences. The CAF-related data for each sample were obtained through three different algorithms. Consistency cluster analysis was performed to obtain different CAF clusters, which were analyzed to identify differentially expressed genes. These were subjected to uniform cluster analysis to obtain different gene clusters. The Boruta algorithm was used to calculate the CAF score. Three CAF clusters and two gene clusters were obtained, all of which differed in their patient prognoses and the content of infiltrating immune cells. Patients with high CAF scores exhibited worse overall survival, higher expression of biomarkers related to immune checkpoints and immune activity, and lower tumor mutation burden. The CAF score could also predict the immunotherapy response of patients. This study suggests that the CAF score can accurately predict the prognosis and immunotherapy response of LUSC patients.
Collapse
Affiliation(s)
- Yinhui Zhu
- Department of Respiratory and Critical Care Medicine, The Third Hospital of Changsha, Hunan, China
| | - Yingqun Zhu
- Department of Respiratory and Critical Care Medicine, The Third Hospital of Changsha, Hunan, China
| | - Sirui Chen
- Department of Emergency Medicine, The Third Hospital of Changsha, Hunan, China
| | - Qian Cai
- Department of Respiratory and Critical Care Medicine, The Third Hospital of Changsha, Hunan, China
| |
Collapse
|
4
|
Dowdell AK, Meng RC, Vita A, Bapat B, Hanes D, Chang SC, Harold L, Wong C, Poon H, Schroeder B, Weerasinghe R, Leidner R, Urba WJ, Bifulco CB, Piening BD. Widespread Adoption of Precision Anticancer Therapies After Implementation of Pathologist-Directed Comprehensive Genomic Profiling Across a Large US Health System. JCO Oncol Pract 2024; 20:1523-1532. [PMID: 39531849 PMCID: PMC11623383 DOI: 10.1200/op.24.00226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/25/2024] [Accepted: 08/16/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE Precision therapies and immunotherapies have revolutionized cancer care, with novel genomic biomarker-associated therapies being introduced into clinical practice rapidly, resulting in notable gains in patient survival. Despite this, there is significant variability in the utilization of tumor molecular profiling that spans the timing of test ordering, comprehensiveness of gene panels, and clinical decision support through therapy and trial recommendations. METHODS To standardize testing, we designed a pathologist-directed test ordering system at the time of diagnosis using a 523-gene DNA/RNA hybrid comprehensive genomic profiling (CGP) panel and extensive clinical decision support tools. To comprehensively characterize the clinical impact of this protocol, we developed a novel natural language processing (NLP)-based approach to extract clinical features from physician chart notes. We assessed test actionability rates, therapy choice, and outcomes across a set of 3,216 patients with advanced cancer. RESULTS We observed 49% of patients had at least one actionable genomic biomarker-driven-approved and/or guideline-recommended targeted or immunotherapy (IO) and 53% of patients would have been eligible for a precision therapy clinical trial from three large basket trials. When assessing CGP versus an in silico 50-gene panel, 67% of tumors compared with 33% harbored actionable alterations including clinical trials. Among patients with 6 months or more of follow-up, over 52% received a targeted therapy (TT) or IO, versus 32% who received conventional chemotherapy alone. Furthermore, patients receiving TT had significantly improved overall survival compared with patients receiving chemotherapy alone (P < .001). CONCLUSION Overall, these data represent a major shift in standard clinical practice toward molecularly guided treatments (targeted and immunotherapies) over conventional systemic chemotherapy. As guidelines continue to evolve and more precision therapeutics gain approval, we expect this gap to continue to widen.
Collapse
Affiliation(s)
- Alexa K. Dowdell
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | - Ryan C. Meng
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | | | | | | | | | - Lauren Harold
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | | | | | | | | | - Rom Leidner
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | - Walter J. Urba
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | - Carlo B. Bifulco
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| | - Brian D. Piening
- Providence Health, Portland, OR
- Earle A. Chiles Research Institute, Portland, OR
| |
Collapse
|
5
|
Huang K, Liu H, Wu Y, Fan W, Zhao Y, Xue M, Tang Y, Feng ST, Li J. Development and validation of survival prediction models for patients with hepatocellular carcinoma treated with transcatheter arterial chemoembolization plus tyrosine kinase inhibitors. LA RADIOLOGIA MEDICA 2024; 129:1597-1610. [PMID: 39400683 DOI: 10.1007/s11547-024-01890-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Due to heterogeneity of molecular biology and microenvironment, therapeutic efficacy varies among hepatocellular carcinoma (HCC) patients treated with transcatheter arterial chemoembolization (TACE) and tyrosine kinase inhibitors (TKIs). We examined combined models using clinicoradiological characteristics, mutational burden of signaling pathways, and radiomics features to predict survival prognosis. METHODS Two cohorts comprising 111 patients with HCC were used to build prognostic models. The training and test cohorts included 78 and 33 individuals, respectively. Mutational burden was calculated based on 17 cancer-associated signaling pathways. Radiomic features were extracted and selected from computed tomography images using a pyradiomics system. Models based on clinicoradiological indicators, mutational burden, and radiomics score (rad-score) were built to predict overall survival (OS) and progression-free survival (PFS). RESULTS Eastern Cooperative Oncology Group performance status, Child-Pugh class, peritumoral enhancement, PI3K_AKT and hypoxia mutational burden, and rad-score were used to create a combined model predicting OS. C-indices were 0.805 (training cohort) and 0.768 (test cohort). The areas under the curve (AUCs) were 0.889, 0.900, and 0.917 for 1-year, 2-year, and 3-year OS, respectively. To predict PFS, alpha-fetoprotein level, tumor enhancement pattern, hypoxia and receptor tyrosine kinase mutational burden, and rad-score were used. C-indices were 0.782 (training cohort) and 0.766 (test cohort). AUCs were 0.885 and 0.925 for 6-month and 12-month PFS, respectively. Calibration and decision curve analyses supported the model's accuracy and clinical potential. CONCLUSIONS The nomogram models are hopeful to predict OS and PFS in patients with intermediate-advanced HCC treated with TACE plus TKIs, offering a promising tool for treatment decisions and monitoring patient progress.
Collapse
Affiliation(s)
- Kun Huang
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, China
- Department of Radiology, Guizhou Provincial People's Hospital, No. 83 East Zhongshan Road, Guiyang, 550002, Guizhou, China
| | - Haikuan Liu
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, China
| | - Yanqin Wu
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, China
| | - Wenzhe Fan
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, China
| | - Yue Zhao
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, China
| | - Miao Xue
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, China
| | - Yiyang Tang
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, China.
| | - Jiaping Li
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
6
|
Wang K, Leyba A, Hsu R. Addressing the unmet need in NSCLC progression with advances in second-line therapeutics. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1297-1320. [PMID: 39759220 PMCID: PMC11700623 DOI: 10.37349/etat.2024.00277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/16/2024] [Indexed: 01/07/2025] Open
Abstract
Lung cancer is the leading cause of cancer mortality globally, with non-small cell lung cancer (NSCLC) accounting for 85% of cases. Despite advancements in first-line treatments such as immunotherapy and targeted therapies, resistance to these treatments is common, creating a significant unmet need for effective second-line therapies. This review evaluates current and emerging second-line therapeutic options for advanced or metastatic NSCLC, focusing on their efficacy and potential to improve patient outcomes. Anti-angiogenic drugs like ramucirumab combined with chemotherapy, particularly docetaxel, have shown moderate success. Antibody-drug conjugates (ADCs) targeting specific tumor antigens offer a promising avenue for targeted therapy, while chimeric antigen receptor (CAR)-T cell therapy and T-cell receptor therapy leverage the patient's immune system to combat cancer more effectively. mRNA vaccines, although in early stages, show potential for inducing robust immune responses against cancer-specific antigens. Building on this foundation, recent advancements in molecular testing and the exploration of the tumor microenvironment are opening new therapeutic avenues, further enhancing the potential for personalized second-line treatments in NSCLC. While ADCs and bispecific antibodies are gaining traction, more precise biomarkers are needed to optimize treatment response. Regular monitoring through techniques like liquid biopsies allows real-time tracking of mutations such as EGFR T790M, enabling timely therapeutic adjustments. Additionally, the role of neutrophils and macrophages in the tumor microenvironment is increasingly being recognized as a potential therapeutic avenue, with Smad3 emerging as a key target. Further research into drug sequencing, toxicity management, and biomarker development remains crucial to improving NSCLC treatment outcomes.
Collapse
Affiliation(s)
- Kinsley Wang
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Alexis Leyba
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Robert Hsu
- Department of Medicine, Division of Medical Oncology, University of Southern California Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| |
Collapse
|
7
|
Ilikci-Sagkan R, Fatma Akin D, Liman R, Muddassir Ali M. In silico analysis of DEL-1 and inflammation-related genes in lung squamous cell carcinoma. Immunobiology 2024; 229:152838. [PMID: 39089131 DOI: 10.1016/j.imbio.2024.152838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024]
Abstract
AIM Twenty to thirty percent of non-small cell lung cancers (NSCLC) are caused by lung squamous cell carcinoma (LUSC), especially in smokers and there has been limited study previously evaluating the situation in terms of the genome and gene expression profile, which demonstrates the relationship among DEL-1, leucocyte recruitment, and pro-inflammatory cytokines in LUSC. MATERIAL AND METHODS In the current study, the m-RNA expression patterns and mutation profiles of our target genes, such as, pro-inflammatory cytokines, chemoattractant molecules, and DEL-1 genes, in 511 LUSC patients. To find the harmful mutations, the PolyPhen-2 and SNAP programs were employed. Not only gene expression was detected, but also survival analysis and correlation between DEL-1 and other target genes' expression levels were explored too. RESULTS Target genes such as, DEL-1, TNF, IL-18, IL-1, CXCL8, CXCL13, and IL-6 were found to have a total genetic anomaly carrying rate of 16.4%. Seven mutations were found, and two of those mutations have a pathogenic aspect. Deep deletion and gene amplification of the genetic anomalies were also observed. According to gene expression analysis results in the LUSC patient group; DEL-1 and IL-6 levels were significantly lower than those of the control group, whereas the CXCL13 level was found to be higher. CONCLUSION Findings of the current study revealed that, there is a significant role of DEL-1 in LUSC pathogenesis. Since present study is an in silico-centered study, this approach can give more insight on experimental studies. These events may support that one of the cancer improvement mechanisms depending on DEL-1 gene at the molecular level.
Collapse
Affiliation(s)
- Rahsan Ilikci-Sagkan
- Uşak University, School of Medicine, Department of Medical Biology, Uşak, Türkiye.
| | - Dilara Fatma Akin
- Nigde Omer Halisdemir University, School of Medicine, Department of Medical Biology, Niğde, Türkiye
| | - Recep Liman
- Uşak University, Faculty of Engineering and Natural Sciences, Department of Molecular Biology and Genetic, Uşak, Türkiye
| | - Muhammad Muddassir Ali
- Institute of Biochemistry and Biotechnology, University of Veterinary and Animal Sciences, Lahore, Pakistan.
| |
Collapse
|
8
|
Sun D, Tan L, Chen Y, Yuan Q, Jiang K, Liu Y, Xue Y, Zhang J, Cao X, Xu M, Luo Y, Xu Z, Xu Z, Xu W, Shen M. CXCL5 impedes CD8 + T cell immunity by upregulating PD-L1 expression in lung cancer via PXN/AKT signaling phosphorylation and neutrophil chemotaxis. J Exp Clin Cancer Res 2024; 43:202. [PMID: 39034411 PMCID: PMC11264977 DOI: 10.1186/s13046-024-03122-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Lung cancer remains one of the most prevalent cancer types worldwide, with a high mortality rate. Upregulation of programmed cell death protein 1 (PD-1) and its ligand (PD-L1) may represent a key mechanism for evading immune surveillance. Immune checkpoint blockade (ICB) antibodies against PD-1 or PD-L1 are therefore widely used to treat patients with lung cancer. However, the mechanisms by which lung cancer and neutrophils in the microenvironment sustain PD-L1 expression and impart stronger inhibition of CD8+ T cell function remain unclear. METHODS We investigated the role and underlying mechanism by which PD-L1+ lung cancer and PD-L1+ neutrophils impede the function of CD8+ T cells through magnetic bead cell sorting, quantitative real-time polymerase chain reaction (RT-PCR), western blotting, enzyme-linked immunosorbent assays, confocal immunofluorescence, gene silencing, flow cytometry, etc. In vivo efficacy and safety studies were conducted using (Non-obeseDiabetes/severe combined immune deficiency) SCID/NOD mice. Additionally, we collected clinical and prognostic data from 208 patients who underwent curative lung cancer resection between 2017 and 2018. RESULTS We demonstrated that C-X-C motif chemokine ligand 5 (CXCL5) is markedly overexpressed in lung cancer cells and is positively correlated with a poor prognosis in patients with lung cancer. Mechanistically, CXCL5 activates the phosphorylation of the Paxillin/AKT signaling cascade, leading to upregulation of PD-L1 expression and the formation of a positive feedback loop. Moreover, CXCL5 attracts neutrophils, compromising CD8+ T cell-dependent antitumor immunity. These PD-L1+ neutrophils aggravate CD8+ T cell exhaustion following lung cancer domestication. Combined treatment with anti-CXCL5 and anti-PD-L1 antibodies significantly inhibits tumor growth in vivo. CONCLUSIONS Our findings collectively demonstrate that CXCL5 promotes immune escape through PD-L1 upregulation in lung cancer and neutrophils chemotaxis through autocrine and paracrine mechanisms. CXCL5 may serve as a potential therapeutic target in synergy with ICBs in lung cancer immunotherapy.
Collapse
Affiliation(s)
- Dantong Sun
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Lipin Tan
- Department of nursing administration, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yongbing Chen
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Qiang Yuan
- Department of interventional medicine, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Kanqiu Jiang
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yangyang Liu
- Department of Vascular Surgery, Hospital of Zhangjiagang, Suzhou, 215600, China
| | - Yuhang Xue
- Department of Thoracic Surgery, Hospital of Yancheng, Yancheng, 224000, China
| | - Jinzhi Zhang
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Xianbao Cao
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Minzhao Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yang Luo
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Zhonghua Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Zhonghen Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Weihua Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Mingjing Shen
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| |
Collapse
|
9
|
Shahin M, Patra S, Purkait S, Kar M, Das Majumdar SK, Mishra TS, Samal SC, Nayak HK. PD-L1 Expression in Colorectal Carcinoma Correlates with the Immune Microenvironment. J Gastrointest Cancer 2024; 55:940-949. [PMID: 38530597 DOI: 10.1007/s12029-024-01049-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 03/28/2024]
Abstract
INTRODUCTION/BACKGROUND Colorectal carcinoma (CRC) is a common malignancy, with its diverse clinical, pathological, and molecular features. The immune microenvironment of a tumor comprises of interplay between various cells and molecules, and has a significant role in deciding the tumor behavior and overall prognosis. PD-L1 (programmed cell death ligand-1) has been implicated in the regulation of the tumor immune microenvironment (TIME). There is limited data regarding the correlation of PD-L1 expression with immune cell profile in CRCs, especially in the Indian setting. The study aimed to assess the PD-L1 expression in CRC tumor cells and its association with TIME, mismatch repair (MMR), and various other clinicopathological parameters. METHODS This is a hospital-based, cross-sectional observational study. PD-L1 expression was assessed at the protein level by immunohistochemistry and mRNA level by qRT-PCR. Immune cell markers (CD4, CD8, CD20, FOXP3, and CD163) were interpreted using the ImageJ Fiji platform. RESULTS Of the 104 cases, 21% were PD-L1 positive and were more common in right-sided CRCs. PD-L1 positive cases showed significantly higher concentrations of all T-cell subsets (CD4+ , CD8+ , and FOXP3+), CD20+ B-cells, and CD163+ macrophages were noted. No statistical significance was seen between PD-L1 expression with clinical profile, pathological subtype, grade or stage, mismatch repair status (proficient vs deficient), and survival. CONCLUSIONS The present study showed a relatively lower frequency of PD-L1 in CRC from the Eastern Indian cohort. The immune cell concentration in the present study was calculated using image analysis-based objectivised methods. Significant correlation of PD-L1 expression in tumor cells with the tumor-infiltrating immune cells indicated its crucial role in the pathobiology of CRC especially by regulating the TIME. Considering the therapeutic implication of PD-L1 in various malignancies, it may be one of the crucial therapeutic targets in a proportion of cases.
Collapse
Affiliation(s)
- Mohammed Shahin
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Susama Patra
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India.
| | - Suvendu Purkait
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Madhabananda Kar
- Department of Surgical Oncology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Saroj Kumar Das Majumdar
- Department of Radiation Oncology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | | | - Subash Chandra Samal
- Department of Medical Gastroenterology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Hemanta Kumar Nayak
- Department of Medical Gastroenterology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| |
Collapse
|
10
|
Liu F, Zhang X, Lu M, Liu C, Zhang X, Chu Q, Chen Y, Zhang P. The association of genomic alterations with PD-L1 expression in Chinese patients with EGFR/ALK wild-type lung adenocarcinoma and potential predictive value of Hippo pathway mutations to immunotherapy. Cancer Med 2024; 13:e7038. [PMID: 38396367 PMCID: PMC10891359 DOI: 10.1002/cam4.7038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/23/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The study focuses on PD-L1 expression as an essential biomarker for gauging the response of EGFR/ALK wild-type NSCLC patients to FDA-approved immune checkpoint inhibitors (ICIs). It aims to explore clinical, molecular, and immune microenvironment characteristics associated with PD-L1 expression in EGFR/ALK wild-type lung adenocarcinoma patients eligible for ICI therapy. METHODS In this retrospective study, tumor samples from 359 Chinese EGFR/ALK wild-type lung adenocarcinoma patients underwent comprehensive evaluations for PD-L1 expression and NGS-targeted sequencing. The investigation encompassed the analysis and comparison of clinical traits, gene mutations, pathways, and immune signatures between two groups categorized by PD-L1 status: negative (TPS < 1%) and positive (TPS ≥ 1%). Additionally, the study explored the link between genomic changes and outcomes following immunotherapy. RESULTS High tumor mutational burden correlated significantly with PD-L1 positivity in patients with EGFR/ALK wild-type lung adenocarcinoma. Gene alterations, including TP53, KRAS, and others, were more pronounced in the PD-L1 positive group. Pathway analysis highlighted higher frequencies of alterations in pathways like RTK/RAS, p53, and Hippo in PD-L1-positive patients. The Hippo pathway's relevance was confirmed in separate immunotherapy cohorts, associated with better outcomes. In terms of immune cell infiltration, Hippo mutants exhibited higher levels of CD68+ PD-L1+ macrophages, CD8+ T cells, and CD8+ PD-1- T cells. CONCLUSIONS This study offers insights into genomic features of Chinese EGFR/ALK wild-type lung adenocarcinoma patients based on PD-L1 expression. Notably, Hippo pathway alterations were linked to improved immunotherapy outcomes. These findings suggest connections between the Hippo pathway and PD-L1 expression, warranting further clinical and functional investigations. The research advances our understanding of PD-L1 expression's genomic context and immunotherapy response in EGFR/ALK wild-type lung adenocarcinoma.
Collapse
Affiliation(s)
- Fangfang Liu
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xuemei Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Mengyao Lu
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chun Liu
- Genecast Biotechnology Co., LtdWuxiJiangsuChina
| | | | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuan Chen
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Peng Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
11
|
Zhang G, Ji P, Xia P, Song H, Guo Z, Hu X, Guo Y, Yuan X, Song Y, Shen R, Wang D. Identification and targeting of cancer-associated fibroblast signature genes for prognosis and therapy in Cutaneous melanoma. Comput Biol Med 2023; 167:107597. [PMID: 37875042 DOI: 10.1016/j.compbiomed.2023.107597] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/15/2023] [Accepted: 10/17/2023] [Indexed: 10/26/2023]
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) play pivotal roles in tumor invasion and metastasis. However, studies on CAF biomarkers in Cutaneous Melanoma (CM) are still scarce. This study aimed to explore the potential CAF biomarkers in CM, propose the potential therapeutic targets, and provide new insights for targeted therapy of CAFs in CM. METHODS We utilized weighted gene co-expression network analysis to identify CAF signature genes in CM, and conducted comprehensive bioinformatics analysis on the CAF risk score established by these genes. Moreover, single-cell sequencing analysis, spatial transcriptome analysis, and cell experiments were utilized for verifying the expression and distribution pattern of signature genes. Furthermore, molecular docking was employed to screen potential target drugs. RESULTS FBLN1 and COL5A1, two crucial CAF signature genes, were screened to establish the CAF risk score. Subsequently, a comprehensive bioinformatic analysis of the CAF risk score revealed that high-risk score group was significantly enriched in pathways associated with tumor progression. Besides, CAF risk score was significantly negatively correlated with clinical prognosis, immunotherapy response, and tumor mutational burden in CM patients. In addition, FBLN1 and COL5A1 were further identified as CAF-specific biomarkers in CM by multi-omics analysis and experimental validation. Eventually, based on these two targets, Mifepristone and Dexamethasone were screened as potential anti-CAFs drugs. CONCLUSION The findings indicated that FBLN1 and COL5A1 were the CAF signature genes in CM, which were associated with the progression, treatment, and prognosis of CM. The comprehensive exploration of CAF signature genes is expected to provide new insight for clinical CM therapy.
Collapse
Affiliation(s)
- Guokun Zhang
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China.
| | - Pengfei Ji
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China.
| | - Peng Xia
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China.
| | - Haoyun Song
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China.
| | - Zhao Guo
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China.
| | - Xiaohui Hu
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China.
| | - Yanan Guo
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China.
| | - Xinyi Yuan
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China.
| | - Yanfeng Song
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China.
| | - Rong Shen
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China.
| | - Degui Wang
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China; NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu, 730000, China.
| |
Collapse
|
12
|
Fulton-Ward T, Middleton G. The impact of genomic context on outcomes of solid cancer patients treated with genotype-matched targeted therapies: a comprehensive review. Ann Oncol 2023; 34:1113-1130. [PMID: 37875224 DOI: 10.1016/j.annonc.2023.10.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/18/2023] [Accepted: 10/08/2023] [Indexed: 10/26/2023] Open
Abstract
INTRODUCTION A critical need in the field of genotype-matched targeted therapy in cancer is to identify patients unlikely to respond to precision medicines. This will manage expectations of individualised therapies and avoid clinical progression to a point where institution of alternative treatments might not be possible. We examined the evidence base of the impact of genomic context on which targeted alterations are inscribed to identify baseline biomarkers distinguishing those obtaining the expected response from those with less benefit from targeted therapies. METHODS A comprehensive narrative review was conducted: scoping searches were undertaken in PubMed, Cochrane Database of Systematic Reviews, and PROSPERO. Outcomes included in meta-analysis were progression-free and overall survival. Data were extracted from Kaplan-Meier and used to calculate hazard ratios. Studies presenting data on two molecular subcohorts (e.g. co-mutation versus no co-mutation) were included in fixed meta-analysis. Other studies were used for descriptive purposes. RESULTS The presence of concomitant driver mutations, higher tumour mutational burden (TMB), greater copy number burden, and APOBEC signatures significantly reduces benefits of targeted therapy in lung cancers in never smokers (LCINS - less than 100 cigarettes per lifetime) and breast cancer, cancers with low TMB. LCINS have significantly poorer outcomes if their cancers harbour p53 co-mutations, an effect also seen in human epidermal growth factor receptor 2-positive (HER2+) breast cancer patients (trastuzumab) and head and neck cancer patients [phosphoinositide 3-kinase (PI3K) inhibition]. PI3K co-alterations have less impact when targeting epidermal growth factor receptor mutations and anaplastic lymphoma kinase fusions, but significantly reduce the impact of targeting HER2 and MET amplifications. SMARCA4 co-mutations predict for poor outcome in patients treated with osimertinib and sotorasib. In BRAF-mutant melanoma, whilst there are no genomic features distinguishing exceptional responders from primary progressors, there are clear transcriptomic features dichotomising these outcomes. CONCLUSION To our knowledge, this is the most comprehensive review to date of the impact of genomic context on outcomes with targeted therapy. It represents a valuable resource informing progress towards contextualised precision medicine.
Collapse
Affiliation(s)
- T Fulton-Ward
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - G Middleton
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; University Hospitals Birmingham, Birmingham, UK.
| |
Collapse
|
13
|
Cheng Y, Chang F, Gong Y, Lu P. A Study on the Clinical Significance of Blood Exosomal PD-L1 in Non-Small Cell Lung Cancer Patients and its Correlation with PD-L1 in Tumor Tissues. Horm Metab Res 2023; 55:788-793. [PMID: 37459865 DOI: 10.1055/a-2110-7497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Exosomal programmed cell-death ligand 1 (ePD-L1) can influence immune inhibition and dysfunction. We were dedicated to unearthing the relation between ePD-L1 in blood and pathological characteristics as well as PD-L1 in tumor tissues. We recruited 65 non-small cell lung cancer (NSCLC) patients for exosome extraction and detected the blood ePD-L1 expression in these patients by enzyme-linked immunosorbent assay (ELISA) method. Besides, the correlation between blood ePD-L1 and patients' pathological characteristics was also analyzed. The expression of PD-L1 in tumor tissues was tested by immunohistochemistry (IHC) and its correlation with blood ePD-L1 expression level was analyzed by Spearman correlation coefficient. No significant correlation was observed in PD-L1 expression levels between blood-derived exosome and tumor tissue. Altogether, high blood ePD-L1 expression was relevant to NSCLC progression, while no such relevance to PD-L1 expression in tumor tissue.
Collapse
Affiliation(s)
- Yonghong Cheng
- Department of Oncology, Shanxi Jishan County People's Hospital, Yuncheng, China
| | - Feiyun Chang
- Department of Thoracic Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Yanmei Gong
- Department of Oncology, Shanxi Yuncheng Center Hospital, Yuncheng, China
| | - Ping Lu
- Department of Radiotherapy Head and Neck Comprehensive Ward, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
14
|
Li W, Zhao Y, Zhang H, Zheng W, Wang R, Gu X. Predictive value of tumor mutational burden for PD-1/PD-L1 inhibitors in NSCLC: A meta-analysis. Medicine (Baltimore) 2023; 102:e34990. [PMID: 37800825 PMCID: PMC10553067 DOI: 10.1097/md.0000000000034990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/08/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND To investigate the association between tumor mutational burden (TMB) and the therapeutic effect of Programmed Death 1/Programmed Death Ligand 1 inhibitors in non-small cell lung cancer. METHODS Four electronic databases, PubMed, Embase, Web of Science, and Cochrane Library, were searched on May 10, 2023, and no time limitation was applied. Analyses were performed using STATA17.0. We assessed the methodological quality of each randomized controlled trial using the Newcastle-Ottawa scale. RESULTS After exhaustive database search and rigorous screening, 10 studies were included in the meta-analysis. Our findings indicate that high TMB significantly improves progression-free survival but reduces overall response rate. The overall survival was not significantly different between the high and low TMB groups. No significant publication bias was observed. CONCLUSION High TMB serves as a potential predictive biomarker for improved progression-free survival and reduced overall response rate in patients with non-small cell lung cancer treated with programmed death 1/programmed death ligand 1 inhibitors. However, its predictive value in overall survival requires further investigation.
Collapse
Affiliation(s)
- Wenjie Li
- Department of Respiratory and Critical Care Medicine, Xi’an Chest Hospital, Chang’an District, Xi’an, Shanxi, China
| | - Yanjun Zhao
- Department of Respiratory and Critical Care Medicine, Xi’an Chest Hospital, Chang’an District, Xi’an, Shanxi, China
| | - Hongjun Zhang
- Department of Respiratory and Critical Care Medicine, Xi’an Chest Hospital, Chang’an District, Xi’an, Shanxi, China
| | - Wenying Zheng
- Department of Respiratory and Critical Care Medicine, Xi’an Chest Hospital, Chang’an District, Xi’an, Shanxi, China
| | - Ruixuan Wang
- Department of Respiratory and Critical Care Medicine, Xi’an Chest Hospital, Chang’an District, Xi’an, Shanxi, China
| | - Xing Gu
- Department of Respiratory and Critical Care Medicine, Xi’an Chest Hospital, Chang’an District, Xi’an, Shanxi, China
| |
Collapse
|
15
|
Wang D, Wu S, He J, Sun L, Zhu H, Zhang Y, Liu S, Duan X, Wang Y, Xu T. FAT4 overexpression promotes antitumor immunity by regulating the β-catenin/STT3/PD-L1 axis in cervical cancer. J Exp Clin Cancer Res 2023; 42:222. [PMID: 37658376 PMCID: PMC10472690 DOI: 10.1186/s13046-023-02758-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 07/10/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND FAT4 (FAT Atypical Cadherin 4) is a member of the cadherin-associated protein family, which has been shown to function as a tumor suppressor by inhibiting proliferation and metastasis. The Wnt/β-catenin pathway activation is highly associated with PD-L1-associated tumor immune escape. Here, we report the mechanism by which FAT4 overexpression regulates anti-tumor immunity in cervical cancer by inhibiting PD-L1 N-glycosylation and cell membrane localization in a β-catenin-dependent manner. METHODS FAT4 expression was first detected in cervical cancer tissues and cell lines. Cell proliferation, clone formation, and immunofluorescence were used to determine the tumor suppressive impact of FAT4 overexpression in vitro, and the findings were confirmed in immunodeficient and immunocomplete mice xenografts. Through functional and mechanistic experiments in vivo and in vitro, we investigated how FAT4 overexpression affects the antitumor immunity via the β-catenin/STT3/PD-L1 axis. RESULTS FAT4 is downregulated in cervical cancer tissues and cell lines. We determined that FAT4 binds to β-catenin and antagonizes its nuclear localization, promotes phosphorylation and degradation of β-catenin by the degradation complexes (AXIN1, APC, GSK3β, CK1). FAT4 overexpression decreases programmed death-ligand 1 (PD-L1) mRNA expression at the transcriptional level, and causes aberrant glycosylation of PD-L1 via STT3A at the post-translational modifications (PTMs) level, leading to its endoplasmic reticulum (ER) accumulation and polyubiquitination-dependent degradation. We found that FAT4 overexpression promotes aberrant PD-L1 glycosylation and degradation in a β-catenin-dependent manner, thereby increasing cytotoxic T lymphocyte (CTL) activity in immunoreactive mouse models. CONCLUSIONS These findings address the basis of Wnt/β-catenin pathway activation in cervical cancer and provide combination immunotherapy options for targeting the FAT4/β-catenin/STT3/PD-L1 axis. Schematic cartoons showing the antitumor immunity mechanism of FAT4. (left) when Wnts bind to their receptors, which are made up of Frizzled proteins and LRP5/6, the cytoplasmic protein DVL is activated, inducing the aggregation of degradation complexes (AXIN, GSK3β, CK1, APC) to the receptor. Subsequently, stable β-catenin translocates into the nucleus and binds to TCF/LEF and TCF7L2 transcription factors, leading to target genes transcription. The catalytically active subunit of oligosaccharyltransferase, STT3A, enhances PD-L1 glycosylation, and N-glycosylated PD-L1 translocates to the cell membrane via the ER-to-Golgi pathway, resulting in immune evasion. (Right) FAT4 exerts antitumor immunity mainly through following mechanisms: (i) FAT4 binds to β-catenin and antagonizes its nuclear localization, promotes phosphorylation and degradation of β-catenin by the degradation complexes (AXIN1, APC, GSK3β, CK1); (ii) FAT4 inhibits PD-L1 and STT3A transcription in a β-catenin-dependent manner and induces aberrant PD-L1 glycosylation and ubiquitination-dependent degradation; (iii) Promotes activation of cytotoxic T lymphocytes (CTL) and infiltration into the tumor microenvironment.
Collapse
Affiliation(s)
- Dongying Wang
- Obstetrics and Gynecology Department, The Second Hospital of Jilin University, 218 Zi Qiang Street, Changchun, Jilin 130041 China
| | - Shuying Wu
- Obstetrics and Gynecology Department, The Second Hospital of Jilin University, 218 Zi Qiang Street, Changchun, Jilin 130041 China
| | - Jiaxing He
- Obstetrics and Gynecology Department, The Second Hospital of Jilin University, 218 Zi Qiang Street, Changchun, Jilin 130041 China
| | - Luguo Sun
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin 130024 China
| | - Hongming Zhu
- Obstetrics and Gynecology Department, The Second Hospital of Jilin University, 218 Zi Qiang Street, Changchun, Jilin 130041 China
| | - Yuxuan Zhang
- Obstetrics and Gynecology Department, The Second Hospital of Jilin University, 218 Zi Qiang Street, Changchun, Jilin 130041 China
| | - Shanshan Liu
- Obstetrics and Gynecology Department, The Second Hospital of Jilin University, 218 Zi Qiang Street, Changchun, Jilin 130041 China
| | - Xuefeng Duan
- Obstetrics and Gynecology Department, The Second Hospital of Jilin University, 218 Zi Qiang Street, Changchun, Jilin 130041 China
| | - Yanhong Wang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin 130024 China
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071 China
| | - Tianmin Xu
- Obstetrics and Gynecology Department, The Second Hospital of Jilin University, 218 Zi Qiang Street, Changchun, Jilin 130041 China
| |
Collapse
|
16
|
Lei X, Li T, Mao F, Ren F, Tang Q, Cao W, Zu L, Xu S. Lobe-specific analysis of perioperative chemotherapy for non-small cell lung cancer patients. Cancer Med 2023; 12:16896-16905. [PMID: 37403701 PMCID: PMC10501251 DOI: 10.1002/cam4.6319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/06/2023] Open
Abstract
OBJECTIVES Perioperative cisplatin-based chemotherapy decreases the risk of death over surgery alone and is a standard of care. Here, we examined perioperative chemotherapy indications for stage IB-III non-small cell lung cancer (NSCLC) patients according to lobe-specific analysis. METHODS Resectable NSCLC patients with stage IB-III who received perioperative chemotherapy with and without radiotherapy after lung resection were identified from the SEER database. Propensity score matching (PSM) analysis was performed to reduce the inherent bias of retrospective studies. The Kaplan-Meier method and log-rank tests were used to assess the differences in overall survival (OS). RESULTS The study enrolled 23,844 patients before PSM. The perioperative chemotherapy group had better OS than the nonperioperative chemotherapy group in stage IB-III NSCLC patients before and after PSM. However, subgroup analysis according to stage demonstrated that perioperative chemotherapy did not markedly benefit patients with stage IB. Furthermore, lobar subgroup analysis did not show survival advantages in primary tumors located in either the right middle lobe in stages II and III NSCLC or the right lower lobe in stage III NSCLC. CONCLUSIONS Lobe-specific perioperative chemotherapy is recommended in NSCLC patients. For stage IB NSCLC, right middle lobe NSCLC from stage IB-III and right lower lobe NSCLC from stage III, perioperative chemotherapy might not confer survival benefits.
Collapse
Affiliation(s)
- Xi Lei
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General HospitalTianjinChina
| | - Tong Li
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General HospitalTianjinChina
| | - Fuling Mao
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General HospitalTianjinChina
| | - Fan Ren
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General HospitalTianjinChina
| | - Quanying Tang
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General HospitalTianjinChina
| | - Weibo Cao
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General HospitalTianjinChina
| | - Lingling Zu
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General HospitalTianjinChina
| | - Song Xu
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General HospitalTianjinChina
| |
Collapse
|
17
|
Hong G, Park HS, Yeo MK, Lee D, Chung C. Dynamic changes in pathology and PD-L1 expression in a patient with metastatic lung adenocarcinoma who received pembrolizumab therapy followed by two salvage surgeries two years later: A case report. Thorac Cancer 2023; 14:2320-2324. [PMID: 37416998 PMCID: PMC10423660 DOI: 10.1111/1759-7714.15023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023] Open
Abstract
During chemotherapy, certain cancer cells undergo cell death, which alters the properties of remaining cells and leads to numerous changes in the constituent cells of lung cancer. Immunotherapy has been used as neoadjuvant therapy, and several studies have reported changes in lung cancer tissue following treatment with immuno-anticancer drugs in early stage disease. However, no research has currently discussed the pathological and PD-L1 expression changes in metastatic lung cancer. Here, we describe a patient with lung adenocarcinoma and multiple metastases who achieved complete remission after receiving initial carboplatin/pemetrexed followed by pembrolizumab treatment for 2-years. The initial biopsy revealed adenocarcinoma with high PD-L1 expression, and next-generation sequencing (NGS) identified KRAS, RBM10, and STAG2 mutations. After 2-years of treatment with pembrolizumab, the patient achieved complete response (CR). The patient underwent first salvage surgery for the oligo-relapse lesion, and the pathology result showed a large cell neuroendocrine tumor (NET) with adenocarcinoma and no PD-L1 expression. NGS revealed KRAS and TP53 mutations. After one year, a chest computed tomography (CT) scan revealed a small nodule in the right lower lobe, and the patient underwent second salvage surgery. Pathology results showed minimally invasive adenocarcinoma with no PD-L1 expression and no significant genetic mutations. This case report demonstrates the dynamic changes cancer cells undergo following pembrolizumab treatment and salvage surgeries and is the first report to compare pathological changes after immunotherapy and two subsequent salvage surgeries in metastatic lung adenocarcinoma. Clinicians must remain vigilant to these dynamic changes throughout treatment and consider salvage surgery for oligo-relapse lesions. By understanding these changes, new strategies can be developed to improve the long-term efficacy of immunotherapy.
Collapse
Affiliation(s)
- Green Hong
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of MedicineChungnam National UniversityDeajeonSouth Korea
| | - Hee Sun Park
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of MedicineChungnam National UniversityDeajeonSouth Korea
| | - Min Kyung Yeo
- Department of Pathology, College of MedicineChungnam National UniversityDeajeonSouth Korea
| | - Dahye Lee
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of MedicineChungnam National UniversityDeajeonSouth Korea
| | - Chaeuk Chung
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of MedicineChungnam National UniversityDeajeonSouth Korea
| |
Collapse
|
18
|
Heenatigala Palliyage G, Samart P, Bobbala S, Rojanasakul LW, Coyle J, Martin K, Callery PS, Rojanasakul Y. Chemotherapy-induced PDL-1 expression in cancer-associated fibroblasts promotes chemoresistance in NSCLC. Lung Cancer 2023; 181:107258. [PMID: 37245409 PMCID: PMC10330668 DOI: 10.1016/j.lungcan.2023.107258] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/30/2023]
Abstract
OBJECTIVES A cure for cancer is out of reach for most patients due to chemoresistance. Cancer-associated fibroblasts (CAFs) play a vital role in cancer chemoresistance, but detailed understanding of the process particularly in chemoresistant lung cancer is lacking. In this study, we investigated programmed death-ligand 1 (PDL-1) as a potential biomarker for CAF-induced chemoresistance and evaluated its role and the underlying mechanisms of chemoresistance in non-small cell lung cancer (NSCLC). MATERIALS AND METHODS A systemic search of gene expression profiles of multiple tissues in NSCLC was carried out to determine the expression intensities of traditional fibroblast biomarkers and CAF-secreted protumorigenic cytokines. PDL-1 expression in CAFs was analyzed by ELISA, Western blotting, and flow cytometry. Human cytokine array was used to identify specific cytokines secreted from CAFs. Role of PDL-1 in NSCLC chemoresistance was assessed using CRISPR/Cas9 knockdown and various functional assays including MTT, cell invasion, sphere formation, and cell apoptosis. In vivo experiments were conducted using a co-implantation xenograft mouse model with live cell imaging and immunohistochemistry. RESULTS We demonstrated that chemotherapy-stimulated CAFs promoted tumorigenic and stem cell-like properties of NSCLC cells, which contribute to their chemoresistance. Subsequently, we revealed that PDL-1 expression is upregulated in chemotherapy-treated CAFs and is associated with poor prognosis. Silencing PDL-1 expression suppressed CAFs' ability to promote stem cell-like properties and invasiveness of lung cancer cells, favoring chemoresistance. Mechanistically, an upregulation of PDL-1 in chemotherapy-treated CAFs led to an increase in hepatocyte growth factor (HGF) secretion, which stimulates cancer progression, cell invasion, and stemness of lung cancer cells, while inhibiting apoptosis. CONCLUSION Our results show that PDL-1-positive CAFs modulate stem cell-like properties of NSCLC cells by secreting elevated HGF, thereby promoting chemoresistance. Our finding supports PDL-1 in CAFs as a chemotherapy response biomarker and as a drug delivery and therapeutic target for chemoresistant NSCLC.
Collapse
Affiliation(s)
| | - Parinya Samart
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA; Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA
| | - Liying W Rojanasakul
- Allergy and Clinical Immunology Branch, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | | | - Karen Martin
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV, USA; West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA; Department of Research & Graduate Education, West Virginia University, Morgantown, WV, USA
| | - Patrick S Callery
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA; West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
19
|
Zheng QM, Li YY, Wang YP, Li GX, Zhao MM, Sun ZG. Association between CD8+ tumor-infiltrating lymphocytes and prognosis of non-small cell lung cancer patients treated with PD-1/PD-L1 inhibitors: a systematic review and meta-analysis. Expert Rev Anticancer Ther 2023; 23:643-659. [PMID: 37114477 DOI: 10.1080/14737140.2023.2208351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
BACKGROUND A meta-analysis method was used to investigate the prognostic value of CD8+ tumor-infiltrating lymphocytes (TILs) in non-small cell lung cancer (NSCLC) patients treated with PD-1/PD-L1 inhibitors. METHODS A database search of PubMed, Embase, Web of Science and Cochrane Library up until February 7th, 2023. A clinical study on the relationship between CD8+ TILs and PD-1/PD-L1 inhibitors in the therapeutics of NSCLC. RevMan 5.3 and StataMP 17.0 software were used for meta-analysis. The outcome indicators incorporated overall survival (OS), progression-free survival (PFS) and objective response rate (ORR). RESULTS Nineteen articles with 1488 patients were included. The analysis results showed that high CD8+ TILs were associated with better OS (HR=0.60, 95% CI: 0.46-0.77; P<0.0001), PFS (HR=0.68, 95% CI: 0.53-0.88; P=0.003) and ORR (OR=2.26, 95% CI: 1.52-3.36; P<0.0001) in NSCLC patients treated with PD-1/PD-L1 inhibitors. Subgroup analysis indicated that patients with high CD8+ TILs had good clinical prognostic benefits whether the location of CD8+ TILs was intratumoral or stromal, and compared with East Asian, high CD8+ TILs in Caucasians showed a better prognosis. High CD8+ TILs in peripheral blood did not improve OS (HR=0.83, 95% CI: 0.69-1.01; P=0.06) and PFS (HR=0.93, 95% CI: 0.61-1.14; P=0.76) in NSCLC patients receiving PD-1/PD-L1 inhibitors. CONCLUSION In spite of the location of CD8+ TILs, high densities of CD8+ TILs were predictive of treatment outcomes in NSCLC patients treated with PD-1/PD-L1 inhibitors. However, high CD8+ TILs in peripheral blood had no predictive effect.
Collapse
Affiliation(s)
- Qi-Ming Zheng
- Department of Thoracic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Yuan-Yuan Li
- Department of Thoracic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013
| | - Ye-Peng Wang
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Guo-Xiang Li
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Meng-Meng Zhao
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Zhi-Gang Sun
- Department of Thoracic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| |
Collapse
|
20
|
Wu S, Zhang Y, Zhang Y, Chen LH, Ouyang HF, Xu X, Du Y, Ti XY. Mutational landscape of homologous recombination-related genes in small-cell lung cancer. Cancer Med 2023; 12:4486-4495. [PMID: 36053931 PMCID: PMC9972032 DOI: 10.1002/cam4.5148] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/27/2022] [Accepted: 08/07/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Homologous recombination deficiency (HRD) is a well-known biomarker which could predict poly-ADP ribose polymerase 1 (PARP) inhibitor and platinum drug response. As an aggressive cancer, small-cell lung cancer (SCLC) is sensitive to platinum drugs, but relapse occurs rapidly. Herein, we aim to illustrate the genomic alteration patterns of homologous recombination repair (HRR)-related genes in a Chinese SCLC cohort and further analyze the relationship among HRR gene mutations and known biomarkers of immune checkpoint inhibitor (ICI) response, including tumor mutation burden (TMB) and programmed cell death-ligand 1 (PD-L1) expression. METHODS Next-generation sequencing (NGS)-based target capture sequencing of 543 cancer-related genes was performed to analyze the genomic profiles of 133 Chinese SCLC patients, and TMB was calculated. PD-L1 expression was evaluated in 90 out of 133 patients using the SP142 PD-L1 immunohistochemistry assay. RESULTS Among the 133 patients with SCLC, 47 (35.3%) had HRR gene mutations. ATM (8.3%) was the most frequently mutated HRR gene in the cohort, followed by NBN (4.5%). Pathogenic somatic and germline mutations of HRR genes were identified in 11 (23.4%) and 4 (8.5%) patients, respectively. HRR gene mutations cooccurred with KMT2D gene mutations. There were several differences in genomic alterations between patients with HRR gene mutations (HRR-Mut) and without HRR mutations (HRR-WT). The results revealed that TP53 and RB1 were commonly mutated genes in both groups. Mutations in the KMT2D gene and genes in the RTK-RAS pathway occurred more frequently in the HRR-Mut group. Furthermore, we found that mutations in HRR genes were associated with high TMB (Wilcoxon, p = 0.048), but there was no correlation of HRR gene mutation status with PD-L1 expression. CONCLUSIONS We exhaustively describe the genomic alteration profile of Chinese SCLC patients and provide further evidence that HRR gene mutations are prevalent in SCLC patients.
Collapse
Affiliation(s)
- Shuo Wu
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi, China
| | - Yao Zhang
- Department of Pulmonary Medicine, Xi'an International Medical Center Hospital, Xi'an, Shanxi, China
| | - Yan Zhang
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi, China
| | - Liz-Han Chen
- Department of Pulmonary Medicine, Xi'an International Medical Center Hospital, Xi'an, Shanxi, China
| | - Hai-Feng Ouyang
- Department of Pulmonary Medicine, Xi'an International Medical Center Hospital, Xi'an, Shanxi, China
| | - Xi Xu
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi, China
| | - Ying Du
- Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu, China
| | - Xin-Yu Ti
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi, China
| |
Collapse
|
21
|
Sadeghirad H, Bahrami T, Layeghi SM, Yousefi H, Rezaei M, Hosseini-Fard SR, Radfar P, Warkiani ME, O'Byrne K, Kulasinghe A. Immunotherapeutic targets in non-small cell lung cancer. Immunology 2023; 168:256-272. [PMID: 35933597 DOI: 10.1111/imm.13562] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 08/02/2022] [Indexed: 01/17/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common types of cancer in the world and has a 5-year survival rate of ~20%. Immunotherapies have shown promising results leading to durable responses, however, they are only effective for a subset of patients. To determine the best therapeutic approach, a thorough and in-depth profiling of the tumour microenvironment (TME) is required. The TME is a complex network of cell types that form an interconnected network, promoting tumour cell initiation, growth and dissemination. The stroma, immune cells and endothelial cells that comprise the TME generate a plethora of cytotoxic or cytoprotective signalling pathways. In this review, we discuss immunotherapeutic targets in NSCLC tumours and how the TME may influence patients' response to immunotherapy.
Collapse
Affiliation(s)
- Habib Sadeghirad
- University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Tayyeb Bahrami
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Sepideh M Layeghi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, Louisiana, USA
| | - Meysam Rezaei
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Seyed R Hosseini-Fard
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Payar Radfar
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Majid E Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Ken O'Byrne
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Arutha Kulasinghe
- University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
22
|
Costimulatory molecule expression profile as a biomarker to predict prognosis and chemotherapy response for patients with small cell lung cancer. Cancer Immunol Immunother 2023; 72:617-631. [PMID: 36002754 PMCID: PMC9947026 DOI: 10.1007/s00262-022-03280-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 08/12/2022] [Indexed: 10/15/2022]
Abstract
Owing to the paucity of specimens, progress in identifying prognostic and therapeutic biomarkers for small cell lung cancer (SCLC) has been stagnant for decades. Considering that the costimulatory molecules are essential elements in modulating immune responses and determining therapeutic response, we systematically revealed the expression landscape and identified a costimulatory molecule-based signature (CMS) to predict prognosis and chemotherapy response for SCLCs for the first time. We found T cell activation was restrained in SCLCs, and costimulatory molecules exhibited widespread abnormal genetic alterations and expression. Using a LASSO Cox regression model, the CMS was built with a training cohort of 77 cases, which successfully divided patients into high- or low-risk groups with significantly different prognosis and chemotherapy benefit (both P < 0.001). The CMS was well validated in an independent cohort containing 131 samples with qPCR data. ROC and C-index analysis confirmed the superior predictive performance of the CMS in comparison with other clinicopathological parameters from different cohorts. Importantly, the CMS was confirmed as a significantly independent prognosticator for clinical outcomes and chemotherapy response in SCLCs through multivariate Cox analysis. Further analysis revealed that low-risk patients were characteristic by an activated immune phenotype with distinct expression of immune checkpoints. In summary, we firstly uncovered the expression heterogeneity of costimulatory molecules in SCLC and successfully constructed a novel predictive CMS. The identified signature contributed to more accurate patient stratification and provided robust prognostic value in estimating survival and the clinical response to chemotherapy, allowing optimization of treatment and prognosis management for patients with SCLC.
Collapse
|
23
|
Xing P, Jiang Z, Liu Y. Construction and validation of a gene signature related to bladder urothelial carcinoma based on immune gene analysis. BMC Cancer 2022; 22:926. [PMID: 36030212 PMCID: PMC9419388 DOI: 10.1186/s12885-022-09794-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 06/15/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND This study developed a gene signature associated with a malignant and common tumor of the urinary system, the Bladder Urothelial Carcinoma (BLCA). METHODS The Cancer Genome Atlas (TCGA) database was searched to obtain 414 BLCA samples and the expression spectra of 19 normal samples. Single-sample Gene Set Enrichment Analysis (ssGSEA) was conducted to determine the enrichment levels in the BLCA samples of the 29 immune genes. Unsupervised hierarchical clustering, gene set enrichment analysis (GSEA), single-factor Cox analysis, least absolute shrinkage and selection operator (LASSO) regression models, and GEO queues were used to determine the BLCA immune gene subtype, analyze the biological pathway differences between immune gene subtypes, determine the characteristic genes of BLCA associated with prognosis, identify the BLCA-related genes, and verify the gene signature, respectively. RESULTS We identified two immune gene subtypes (immunity_L and immunity_H). The latter was significantly related to receptors, JAK STAT signaling pathways, leukocyte interleukin 6 generation, and cell membrane signal receptor complexes. Four characteristic genes (RBP1, OAS1, LRP1, and AGER) were identified and constituted the gene signature. Significant survival advantages, higher mutation frequency, and superior immunotherapy were observed in the low-risk group patients. The gene signature had good predictive ability. The results of the validation group were consistent with TCGA queue results. CONCLUSIONS We constructed a 4-gene signature that helps monitor BLCA occurrence and prognosis, providing an important basis for developing personalized BLCA immunotherapy.
Collapse
Affiliation(s)
- Peng Xing
- Department of Urology, The First Hospital of China Medical University, Shenyang, 110013, P.R. China
| | - Zhengming Jiang
- Department of Urology, The First Hospital of China Medical University, Shenyang, 110013, P.R. China
| | - Yang Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, 110013, P.R. China.
| |
Collapse
|
24
|
Multi-Omics Approaches for the Prediction of Clinical Endpoints after Immunotherapy in Non-Small Cell Lung Cancer: A Comprehensive Review. Biomedicines 2022; 10:biomedicines10061237. [PMID: 35740259 PMCID: PMC9219996 DOI: 10.3390/biomedicines10061237] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized the management of locally advanced and advanced non-small lung cancer (NSCLC). With an improvement in the overall survival (OS) as both first- and second-line treatments, ICIs, and especially programmed-death 1 (PD-1) and programmed-death ligands 1 (PD-L1), changed the landscape of thoracic oncology. The PD-L1 level of expression is commonly accepted as the most used biomarker, with both prognostic and predictive values. However, even in a low expression level of PD-L1, response rates remain significant while a significant number of patients will experience hyperprogression or adverse events. The dentification of such subtypes is thus of paramount importance. While several studies focused mainly on the prediction of the PD-L1 expression status, others aimed directly at the development of prediction/prognostic models. The response to ICIs depends on a complex physiopathological cascade, intricating multiple mechanisms from the molecular to the macroscopic level. With the high-throughput extraction of features, omics approaches aim for the most comprehensive assessment of each patient. In this article, we will review the place of the different biomarkers (clinical, biological, genomics, transcriptomics, proteomics and radiomics), their clinical implementation and discuss the most recent trends projecting on the future steps in prediction modeling in NSCLC patients treated with ICI.
Collapse
|
25
|
Correlation of PD-L1 Expression with Clinicopathological and Genomic Features in Chinese Non-Small-Cell Lung Cancer. JOURNAL OF ONCOLOGY 2022; 2022:1763778. [PMID: 35444698 PMCID: PMC9015849 DOI: 10.1155/2022/1763778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/14/2022] [Indexed: 11/17/2022]
Abstract
Programmed cell death 1 ligand 1 (PD-L1) has been approved as predictive biomarker for non-small-cell lung cancer (NSCLC) patients treated with PD-(L)1 blockade therapy. The clinical/genomic features associated with PD-L1 are not well studied. Genomic profiling of tumor biopsies from 883 Chinese NSCLC patients was performed by targeted next-generation sequencing. Immunohistochemical analysis was conducted to evaluate PD-L1 expression levels using antibodies Dako 22C3 and 28-8, respectively. Our study showed distinct correlation between PD-L1 expression and clinical/genomic characteristics when using different PD-L1 antibodies and in different histological subtypes including adenocarcinoma (ADC) and squamous cell carcinoma (SCC), respectively. PD-L1 high expression (22C3) was associated with male and lymph node metastasis only in ADC patients. Furthermore, mutations of TP53 and KRAS, KIF5B-RET fusion, copy number gains of PD-L1 and PD-L2, and arm-level amplifications of chr.12p were significantly associated with PD-L1 positive status in ADC patients. For SCC patients, the gain of EGFR and MDM2 and loss of PTPRD were negatively associated with PD-L1 expression. We also compared our results with other studies and found conflicting results presumably because of the multiplicity of antibody clones and platforms, the difference of cutoffs for assigning PD-L1 expression levels, and the variation in study populations. Our study can help to understand the utility and validity of PD-L1 as biomarker of response to immune checkpoint inhibitors.
Collapse
|
26
|
Hu ZY, Zheng C, Yang J, Ding S, Tian C, Xie N, Xue L, Wu M, Fu S, Rao Z, Price MA, McCarthy JB, Ouyang Q, Lin J, Deng X. Co-Expression and Combined Prognostic Value of CSPG4 and PDL1 in TP53-Aberrant Triple-Negative Breast Cancer. Front Oncol 2022; 12:804466. [PMID: 35280756 PMCID: PMC8907582 DOI: 10.3389/fonc.2022.804466] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/31/2022] [Indexed: 12/20/2022] Open
Abstract
Background In triple-negative breast cancer (TNBC), PDL1/PD1-directed immunotherapy is effective in less than 20% of patients. In our preliminary study, we have found CSPG4 to be highly expressed together with PDL1 in TNBCs, particularly those harboring TP53 aberrations. However, the clinical implications of co-expressed CSPG4 and PDL1 in TNBCs remain elusive. Methods A total of 85 advanced TNBC patients treated in the Hunan Cancer Hospital between January 2017 and August 2019 were recruited. The expressions of CSPG4 and PDL1 in TNBC tissues were investigated using immunohistochemistry (IHC). The RNA-seq dataset from the TCGA-BRCA project was further used to analyze the mRNA expression of CSPG4 and PDL1 in TP53-aberrant TNBCs. Cox proportional hazards model and Kaplan-Meier curves with Logrank test was used to analyze the effects of CSPG4 and PDL1 on survival. TNBC cell lines were further used to investigate the molecular mechanism that were involved. Results TP53 aberrations occurred in more than 50% of metastatic TNBCs and were related to higher tumor mutation burden (TMB). In TCGA-BRCA RNA-seq dataset analysis, both CSPG4 and PDL1 levels were high in TNBCs, especially in TP53-aberrant TNBCs. IHC assay showed nearly 60% of advanced TNBCs to be CSPG4-positive and about 25% to be both CSPG4-positive and PDL1-positive. The levels of CSPG4 and PDL1 were high in TNBC cell lines as revealed by flow cytometry and immunoblotting compared with non-TNBC cells. Univariate Cox regression analysis indicated that CSPG4 positivity was a significant risk factor for progression-free survival in metastatic TNBCs, with a hazard ratio (HR) of 2.26 (P = 0.05). KM curves with Logrank test also identified high level of CSPG4 as a significant risk factor for overall survival in advanced breast cancers in TCGA-BRCA samples (P = 0.02). The immunoblotting assays showed that EMT-related pathways were involved in CSPG4-mediated invasion. Conclusions CSPG4 expression level is associated with PDL1 positivity in TP53-aberrant TNBC cells. Patients with CSPG4 expression have poor treatment response and poor overall survival. Co-expressed CSPG4 and PDL1 may have an important prognostic value and provide new therapeutic targets in TNBC patients. CSPG4 might mediate tumor invasion and PDL1 overexpression through EMT-related pathway.
Collapse
Affiliation(s)
- Zhe-Yu Hu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China.,Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, China.,Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Chanjuan Zheng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Jianbo Yang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China.,Department of Laboratory Medicine and Pathology and Comprehensive Cancer Center, University of Minnesota, Minneapolis, MN, United States.,The Cancer Center, Union Hospital, Fujian Medical Center, Fuzhou, China
| | - Siyu Ding
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Can Tian
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China.,Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, China.,Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Ning Xie
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China.,Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, China.,Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Lian Xue
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Muyao Wu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Shujun Fu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Zhouzhou Rao
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Matthew A Price
- Department of Laboratory Medicine and Pathology and Comprehensive Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - James B McCarthy
- Department of Laboratory Medicine and Pathology and Comprehensive Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Quchang Ouyang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China.,Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, China.,Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Jizhen Lin
- The Cancer Center, Union Hospital, Fujian Medical Center, Fuzhou, China.,Department of Otolaryngology, Cancer Center, University of Minnesota Medical School, Minnesota, MN, United States
| | - Xiyun Deng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| |
Collapse
|
27
|
Cristescu R, Aurora-Garg D, Albright A, Xu L, Liu XQ, Loboda A, Lang L, Jin F, Rubin EH, Snyder A, Lunceford J. Tumor mutational burden predicts the efficacy of pembrolizumab monotherapy: a pan-tumor retrospective analysis of participants with advanced solid tumors. J Immunother Cancer 2022; 10:jitc-2021-003091. [PMID: 35101941 PMCID: PMC8804694 DOI: 10.1136/jitc-2021-003091] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Several studies have evaluated the relationship between tumor mutational burden (TMB) and outcomes of immune checkpoint inhibitors. In the phase II KEYNOTE-158 study of pembrolizumab monotherapy for previously treated recurrent or metastatic cancer, high TMB as assessed by the FoundationOne CDx was associated with an improved objective response rate (ORR). METHODS We retrospectively assessed the relationship between TMB and efficacy in participants with previously treated advanced solid tumors enrolled in 12 trials that evaluated pembrolizumab monotherapy, including 3 randomized trials that compared pembrolizumab with chemotherapy. TMB was assessed in formalin-fixed, paraffin-embedded pretreatment tumor samples by whole-exome sequencing. High TMB was defined as ≥175 mutations/exome. Microsatellite instability (MSI) phenotype was based on whole-exome sequencing results. Programmed death ligand 1 (PD-L1) expression was assessed by immunohistochemistry. The primary end point was ORR assessed per RECIST V.1.1 by independent central review. Other end points included progression-free survival (PFS) assessed per RECIST V.1.1 by independent central review and overall survival (OS). RESULTS Of the 2234 participants in the analysis, 1772 received pembrolizumab monotherapy and 462 received chemotherapy. Among the pembrolizumab-treated participants, ORR was 31.4% (95% CI 27.1 to 36.0) in the 433 participants with TMB ≥175 mutations/exome and 9.5% (95% CI 8.0 to 11.2) in the 1339 participants with TMB <175 mutations/exome. The association of TMB with ORR was observed regardless of PD-L1 expression and not driven by specific tumor types or participants with very high TMB or high MSI. In the 3 randomized controlled trials, TMB was associated with ORR (p≤0.016), PFS (p≤0.005), and OS (p≤0.029) of pembrolizumab but not of chemotherapy (p≥0.340, p≥0.643, and p≥0.174, respectively), and pembrolizumab improved efficacy versus chemotherapy in participants with TMB ≥175 mutations/exome. CONCLUSIONS TMB ≥175 mutations/exome is associated with clinically meaningful improvement in the efficacy of pembrolizumab monotherapy and improved outcomes for pembrolizumab versus chemotherapy across a wide range of previously treated advanced solid tumor types. These data suggest TMB has broad clinical utility irrespective of tumor type, PD-L1 expression, or MSI status and support its use as a predictive biomarker for pembrolizumab monotherapy in participants with previously treated advanced solid tumors.
Collapse
Affiliation(s)
| | | | | | - Lei Xu
- Merck & Co, Inc, Kenilworth, New Jersey, USA
| | | | | | - Lixin Lang
- Merck & Co, Inc, Kenilworth, New Jersey, USA
| | - Fan Jin
- Merck & Co, Inc, Kenilworth, New Jersey, USA
| | | | | | | |
Collapse
|
28
|
The modulation of PD-L1 induced by the oncogenic HBXIP for breast cancer growth. Acta Pharmacol Sin 2022; 43:429-445. [PMID: 33824459 PMCID: PMC8791967 DOI: 10.1038/s41401-021-00631-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/24/2021] [Indexed: 02/03/2023]
Abstract
Programmed death ligand-1 (PD-L1)/PD-1 checkpoint extensively serves as a central mediator of immunosuppression. A tumor-promoting role for abundant PD-L1 in several cancers is revealed. However, the importance of PD-L1 and how the PD-L1 expression is controlled in breast cancer remains obscure. Here, the mechanisms of controlling PD-L1 at the transcription and protein acetylation levels in promoting breast cancer growth are presented. Overexpressed PD-L1 accelerates breast cancer growth in vitro and in vivo. RNA-seq uncovers that PD-L1 can induce some target genes affecting many cellular processes, especially cancer development. In clinical breast cancer tissues and cells, PD-L1 and HBXIP are both increased, and their expressions are positively correlated. Mechanistic exploration identifies that HBXIP stimulates the transcription of PD-L1 through co-activating ETS2. Specifically, HBXIP induces PD-L1 acetylation at K270 site through interacting with acetyltransferase p300, leading to the stability of PD-L1 protein. Functionally, depletion of HBXIP attenuates PD-L1-accelerated breast tumor growth. Aspirin alleviates breast cancer via targeting PD-L1 and HBXIP. Collectively, the findings display new light into the mechanisms of controlling tumor PD-L1 and broaden the utility for PD-L1 as a target in breast cancer therapy.
Collapse
|
29
|
Wang Y, Gu W, Wen W, Zhang X. SERPINH1 is a Potential Prognostic Biomarker and Correlated With Immune Infiltration: A Pan-Cancer Analysis. Front Genet 2022; 12:756094. [PMID: 35058967 PMCID: PMC8764125 DOI: 10.3389/fgene.2021.756094] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/06/2021] [Indexed: 01/14/2023] Open
Abstract
Background: Serpin peptidase inhibitor clade H, member 1 (SERPINH1) is a gene encoding a member of the serpin superfamily of serine proteinase inhibitors. The upregulated of SERPINH1 was associated with poor prognosis in breast cancer, stomach adenocarcinoma, and esophageal carcinoma. However, the role of SERPINH1 in pan-cancer is largely unexplored. Methods: SERPINH1 expression and the correlation with prognosis in human pan-cancer were analyzed by the Cancer Genome Atlas and the Genotype-Tissue Expression dataset. Pearson correlation analysis was applied to evaluate the role of SERPINH1 expression in tumor mutation burden (TMB), microsatellite instability (MSI), mismatch repair (MMR), DNA methyltransferase, and common immunoregulators. Spearman’s correlation test was used to analysis SERPINH1 expression in tumor immune infiltration and infiltrating immune cells via the Tumor Immune Evaluation Resource database. Furtherly, immunohistochemistry staining of SERPINH1 was acquired from the Human Protein Atlas database for validation. Results: SERPINH1 was abnormally expressed in fourteen cancers. The high expression of SERPINH1 significantly reduced the overall survival (OS), disease-specific survival, and progression free interval in eleven cancers. Moreover, SERPINH1 expression was correlated with MMR, MSI, TMB, and DNA methylation in multiple types of cancer. Also, SERPINH1 expression showed strong association with immunoregulators and immune checkpoint markers in testicular germ cell tumors, brain lower grade glioma (LGG), pheochromocytoma and paraganglioma. In addition, SERPINH1 expression was related to immune cell infiltration in multiple cancers, particularly in breast invasive carcinoma, LGG, and liver hepatocellular carcinoma. The result of immunohistochemistry verification shown that SERPINH1 staining was higher in tumor samples than in normal tissue in colon adenocarcinoma, head and neck squamous cell carcinoma, kidney renal papillary cell carcinoma and cervical squamous cell carcinoma, which was consistent with the result of OS. Conclusion: Overall, these results indicate that SERPINH1 may serve as an important prognostic biomarker and correlate with tumor immunity in human pan-cancer.
Collapse
Affiliation(s)
- Yu Wang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Hangzhou Institute of Digestive Diseases, Hangzhou, China.,Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Weigang Gu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Hangzhou Institute of Digestive Diseases, Hangzhou, China.,Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Weiwei Wen
- Department of Dermatology, Third People's Hospital of Hangzhou, Hangzhou, China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Hangzhou Institute of Digestive Diseases, Hangzhou, China.,Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
30
|
Zou S, Ye J, Hu S, Wei Y, Xu J. Mutations in the TTN Gene are a Prognostic Factor for Patients with Lung Squamous Cell Carcinomas. Int J Gen Med 2022; 15:19-31. [PMID: 35018111 PMCID: PMC8742622 DOI: 10.2147/ijgm.s343259] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/20/2021] [Indexed: 01/04/2023] Open
Abstract
Purpose To analyze the relationship between titin (TTN) mutation gene and tumor mutational burden (TMB) and the with prognosis of lung squamous cell carcinomas (LUSC), and to explore the feasibility of TTN as a potential prognostic marker of for LUSC. Methods We analyzed the somatic mutation landscape of LUSC samples using datasets obtained from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. Sequence data were divided into wild and mutant groups, and differences in TMB values between the groups compared using a Mann–Whitney U-test. The Kaplan Meier method was used to analyze the correlation between TTN mutation and LUSC prognosis, whereas CIBERSORT algorithm was used to calculate the degree of relative enrichment degree of among tumor-infiltrating lymphocytes in LUSC. Results Analysis of both datasets revealed high mutations in the TTN gene, with mutants exhibiting a significantly higher TMB value relative to the wild-type (P < 0.001). Prognosis of the TTN mutant group in LUSC was significantly better than that of wild-type (P = 0.009). Kaplan Meier curves showed that TTN mutation may be an independent prognostic factor in LUSC patients (HR: 0.64, 95% CI 0.48–0.85, P = 0.001), while GSEA analysis revealed that TTN mutation plays a potential role in the development of LUSC. Finally, analysis of LUSC immune microenvironment revealed that TTN mutation was significantly associated with enrichment of macrophages M1 (p < 0.05). Conclusion TTN mutation is associated with TMB, and is positively correlated with prognosis of LUSC. Therefore, this mutation may serve as a potential prognostic indicator of LUSC.
Collapse
Affiliation(s)
- Sheng Zou
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Jiayue Ye
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Sheng Hu
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Yiping Wei
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Jianjun Xu
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, People's Republic of China
| |
Collapse
|
31
|
Li Y, Meng Y, Sun H, Ye L, Zeng F, Chen X, Deng G. The Prognostic Significance of Baseline Neutrophil-to-Lymphocyte Ratio in Melanoma Patients Receiving Immunotherapy. J Immunother 2022; 45:43-50. [PMID: 34510106 PMCID: PMC8654256 DOI: 10.1097/cji.0000000000000392] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Immunotherapy has revolutionized the treatment in metastatic melanoma, but alternative biomarkers that are economical, simple and reliable still need to be clarified. In this study, we aimed to comprehensively analyze the prognostic significance of baseline neutrophil-to-lymphocyte ratio (NLR) in melanoma patients with immunotherapy. We searched PubMed, Embase, and Cochrane Library until September 16, 2020. Hazard ratio (HR) and 95% confidence intervals (CIs) were pooled to investigate the association of baseline NLR with overall survival (OS) and progression-free survival (PFS). Sensitivity analysis, subgroup analyses, publication bias assessment, and the Duval and Tweedie trim-and-fill method were used to evaluate the stability of results. A total of 18 studies including 2054 patients were included in our analysis. Pooled data demonstrated that higher baseline NLR was associated with a poorer OS (HR=2.46, 95% CI=1.77, 3.43) and PFS (HR=2.38, 95% CI=1.95, 2.89) of melanoma patients receiving immunotherapy. Subgroup analysis according to immunotherapy type showed that the prognostic effects of baseline NLR existed in all the subtypes of immunotherapy, including anticytotoxic T lymphocyte-associated protein 4 therapy (OS HR=2.26, 95% CI=1.43, 3.59; PFS HR=2.68, 95% CI=1.79, 4.02), antiprogrammed cell death-1 therapy (OS HR=3.08, 95% CI=2.21, 4.27; PFS HR=2.01, 95% CI=1.64, 2.47), and combination therapy (OS HR=1.75, 95% CI=1.13, 2.72; PFS HR=3.13, 95% CI=1.63, 6.03). Conclusions were still consistent in subgroup analyses stratified by study year, region, study type, sample size, analysis of HR and cuttoff of baseline NLR. Altogether, baseline NLR is a promising prognostic biomarker for melanoma patients receiving immunotherapy.
Collapse
Affiliation(s)
- Yayun Li
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease
- National Clinical Research Center for Geriatric Disorders
| | - Yu Meng
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease
- National Clinical Research Center for Geriatric Disorders
| | - Huiyan Sun
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease
- National Clinical Research Center for Geriatric Disorders
| | - Lin Ye
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease
- National Clinical Research Center for Geriatric Disorders
| | - Furong Zeng
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease
- National Clinical Research Center for Geriatric Disorders
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiang Chen
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease
- National Clinical Research Center for Geriatric Disorders
| | - Guangtong Deng
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease
- National Clinical Research Center for Geriatric Disorders
| |
Collapse
|
32
|
Mao Z, Jiang P, Zhang Y, Li Y, Jia X, Wang Q, Jiao M, Jiang L, Shen Y, Guo H. First-line immune-based combination therapies for advanced non-small cell lung cancer: A Bayesian network meta-analysis. Cancer Med 2021; 10:9139-9155. [PMID: 34747149 PMCID: PMC8683544 DOI: 10.1002/cam4.4405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Immune-based combination therapies have revolutionized the first-line treatment for advanced non-small cell lung cancer (NSCLC). However, for the efficacy and safety, the best treatment option is still uncertain. METHODS We conducted a Bayesian network meta-analysis of randomized controlled trials (RCTs) to evaluate first-line immune-based combination therapies for advanced NSCLC. RESULTS Fourteen trials involving 8467 patients were included. For the programmed cell death-ligand 1 (PD-L1) expression non-selective patients, there were no significant differences among all the treatment modes for overall survival (OS), but the ranking profiles indicated that Immunotherapy + Immunotherapy + Chemotherapy (IO + IO + Chemo) was most likely to be the best mode (probability = 68%). Immunotherapy + Immunotherapy + Anti-angiogenic therapy + Chemotherapy (IO + Anti-angio + Chemo) was significantly better than most other treatment modes for progression-free survival (PFS) with better objective response rate (ORR) and more obvious grade ≥3 treatment-related adverse events (TRAEs). In PD-L1-high cohort, IO + Anti-angio + Chemo seemed to be the best mode for OS, PFS, and ORR according to the ranking profiles. In PD-L1-intermediate and PD-L1-negative cohort, IO + IO + Chemo was inclined to be ranked first for prolonging OS (probability = 78%; 37%) and IO + Anti-angio + Chemo was most likely to provide best PFS (probability = 96%; 100%). CONCLUSION IO + IO + Chemo has great potential to improve the OS regardless of histology type, especially in PD-L1-intermediate and PD-L1-negative cohort. IO + Anti-angio + Chemo shows great superiority in improving the short-term survival accompanied by increasing grade ≥3 TRAEs.
Collapse
Affiliation(s)
- Ziyang Mao
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiP.R. China
| | - Panpan Jiang
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiP.R. China
| | - Yajuan Zhang
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiP.R. China
| | - Yanlin Li
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiP.R. China
| | - Xiaohui Jia
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiP.R. China
| | - Qinyang Wang
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiP.R. China
| | - Min Jiao
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiP.R. China
| | - Lili Jiang
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiP.R. China
| | - Yuan Shen
- Department of Epidemiology and BiostatisticsSchool of Public HealthXi'an Jiaotong University Health Science CenterXi’anShaanxiP.R. China
| | - Hui Guo
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiP.R. China
- Key Laboratory of Environment and Genes Related to DiseasesXi'an Jiaotong UniversityMinistry of Education of ChinaXi'anShaanxiP.R. China
- Centre for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiP.R. China
| |
Collapse
|
33
|
Sun Y, Li L, Yao W, Liu X, Yang Y, Ma B, Xue D. USH2A Mutation is Associated With Tumor Mutation Burden and Antitumor Immunity in Patients With Colon Adenocarcinoma. Front Genet 2021; 12:762160. [PMID: 34795697 PMCID: PMC8593250 DOI: 10.3389/fgene.2021.762160] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/14/2021] [Indexed: 12/26/2022] Open
Abstract
Colon adenocarcinoma (COAD) is one of the diseases with the highest morbidity and mortality in the world. At present, immunotherapy has become a valuable method for the treatment of COAD. Tumor mutational burden (TMB) is considered to be the most common biomarker for predicting immunotherapy. According to reports, the mutation rate of COAD ranks third. However, whether these gene mutations are related to TMB and immune response is still unknown. Here, COAD somatic mutation data were downloaded from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. Bioinformatics methods were used to study the relationships among gene mutations, COAD survival prognosis, and tumor immune response. A total of 22 of the top 40 mutations in TCGA and ICGC databases were the same. Among them, the USH2A mutation was associated with high TMB and poor clinical prognosis. According to Gene Set Enrichment Analysis (GSEA) and the CIBERSORT algorithm, we determined that the USH2A mutation upregulates signaling pathways involved in the immune system and the antitumor immune response. In cases with a USH2A mutation, the immune score and MSI score of TCGA samples increased, the expression of immune checkpoint genes decreased significantly, and the TIDE score decreased significantly. Dependent on the presence or absence of a USH2A mutation, TCGA COAD samples were analyzed for differentially expressed genes, 522 of which were identified. Using a univariate Cox analysis and LASSO COX analysis of these differential genes, a prediction model was established, which established significant differences in the infiltration of immune cells, immune checkpoint gene expression, immune score, MSI score, TMB, and TIDE in patients in high- and low-risk groups. In conclusion, mutation of USH2A is frequent in COAD and is related to an increase in TMB and the antitumor immunity. The differential genes screened by USH2A mutation allowed the construction of a risk model for predicting the survival and prognosis of cancer patients, in addition to providing new ideas for COAD immunotherapy.
Collapse
Affiliation(s)
- Yuanyuan Sun
- Laboratory of Hepatosplenic Surgery, Department of General Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Long Li
- Laboratory of Hepatosplenic Surgery, Department of General Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenchao Yao
- Laboratory of Hepatosplenic Surgery, Department of General Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuxu Liu
- Laboratory of Hepatosplenic Surgery, Department of General Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Yang
- Laboratory of Hepatosplenic Surgery, Department of General Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Biao Ma
- Laboratory of Hepatosplenic Surgery, Department of General Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongbo Xue
- Laboratory of Hepatosplenic Surgery, Department of General Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
34
|
Lu N, Liu J, Xu M, Liang J, Wang Y, Wu Z, Xing Y, Diao F. CSMD3 is Associated with Tumor Mutation Burden and Immune Infiltration in Ovarian Cancer Patients. Int J Gen Med 2021; 14:7647-7657. [PMID: 34764678 PMCID: PMC8575319 DOI: 10.2147/ijgm.s335592] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/28/2021] [Indexed: 12/13/2022] Open
Abstract
Background Globally, ovarian cancer (OC), the deadliest gynecologic malignancy, remains a major cause of mortality, with a rising number of cases in many low- and middle-income countries. Immunotherapy has been proven to be promising for OC. There is increasing awareness of the vital role that tumor mutation burden (TMB) plays in predicting the efficacy of immunotherapy. Women with a family history of OC are at higher risk of the disease due to gene mutations. However, whether these gene mutations are related to immune response and TMB remains to be explored. Methods Our present work analyzed genetic mutation data of OC patients obtained from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) cohorts, and we identified 11 frequently mutated genes, namely, APOB, CSMD3, DST, FAT3, FLG, HMCN1, MUC16, RYR1, TP53, TTN, and USH2A, in accordance with the overlap of two databases. Results A statistically higher TMB was detected by whole-exome sequencing in patients with OC with CSMD3 mutation than in those with mutations in the other frequently mutated genes. Prognosis analysis performed with patients from the TCGA cohort revealed that those with CSMD3 mutation had an overall survival (OS) that was inferior to that of those with wild-type CSMD3. Gene set enrichment analysis (GSEA) and CIBERSORT analysis indicated that OC samples with CSMD3 mutations had significant involvement of pathways related to the immune response. Conclusion In summary, we found that CSMD3 mutation is highly correlated with increased TMB and poor clinical prognosis and that it might function as a biomarker for predicting prognosis and choosing an immunotherapy regimen.
Collapse
Affiliation(s)
- Nan Lu
- Department of Reproduction, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Mengting Xu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Jianqiang Liang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Yichun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Zhipeng Wu
- Department of Urology, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, People's Republic of China
| | - Yan Xing
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Feiyang Diao
- Department of Reproduction, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
35
|
Ma W, Li W, Xu L, Liu L, Xia Y, Yang L, Da M. Identification of a Gene Prognostic Model of Gastric Cancer Based on Analysis of Tumor Mutation Burden. Pathol Oncol Res 2021; 27:1609852. [PMID: 34566519 PMCID: PMC8460769 DOI: 10.3389/pore.2021.1609852] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/27/2021] [Indexed: 01/06/2023]
Abstract
Introduction: Gastric cancer is one of the most common cancers. Although some progress has been made in the treatment of gastric cancer with the improvement of surgical methods and the application of immunotherapy, the prognosis of gastric cancer patients is still unsatisfactory. In recent years, there has been increasing evidence that tumor mutational load (TMB) is strongly associated with survival outcomes and response to immunotherapy. Given the variable response of patients to immunotherapy, it is important to investigate clinical significance of TMB and explore appropriate biomarkers of prognosis in patients with gastric cancer (GC). Material and Methods: All data of patients with gastric cancer were obtained from the database of The Cancer Genome Atlas (TCGA). Samples were divided into two groups based on median TMB. Differently expressed genes (DEGs) between the high- and low-TMB groups were identified and further analyzed. We identified TMB-related genes using Lasso, univariate and multivariate Cox regression analysis and validated the survival result of 11 hub genes using Kaplan-Meier Plotter. In addition, “CIBERSORT” package was utilized to estimate the immune infiltration. Results: Single nucleotide polymorphism (SNP), C > T transition were the most common variant type and single nucleotide variant (SNV), respectively. Patients in the high-TMB group had better survival outcomes than those in the low-TMB group. Besides, eleven TMB-related DEGs were utilized to construct a prognostic model that could be an independent risk factor to predict the prognosis of patients with GC. What’s more, the infiltration levels of CD4+ memory-activated T cells, M0 and M1 macrophages were significantly increased in the high-TMB group compared with the low-TMB group. Conclusions: Herein, we found that patients with high TMB had better survival outcomes in GC. In addition, higher TMB might promote immune infiltration, which could provide new ideas for immunotherapy.
Collapse
Affiliation(s)
- Weijun Ma
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, China
| | - Weidong Li
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, China
| | - Lei Xu
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, China.,The First Clinical Medical College, Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Lu Liu
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, China.,The First Clinical Medical College, Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Yu Xia
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, China.,First Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Liping Yang
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, China
| | - Mingxu Da
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
36
|
Cho YA, Lee H, Kim DG, Kim H, Ha SY, Choi YL, Jang KT, Kim KM. PD-L1 Expression Is Significantly Associated with Tumor Mutation Burden and Microsatellite Instability Score. Cancers (Basel) 2021; 13:cancers13184659. [PMID: 34572886 PMCID: PMC8466224 DOI: 10.3390/cancers13184659] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Biomarkers for predicting the response to immune checkpoint blockade (ICB) includes programmed death-ligand 1 (PD-L1) immunohistochemistry (IHC), microsatellite instability (MSI), and tumor mutation burden (TMB). This study investigated the relationship of these biomarkers using comprehensive cancer panel assay (CCPA) with >500 genes in 588 advanced cancer patients. The work demonstrates that PD-L1 expression is significantly associated with TMB and MSI score, according to primary tumor origin. Abstract Programmed death-ligand 1 (PD-L1) immunohistochemistry (IHC), microsatellite instability (MSI), and tumor mutation burden (TMB) have been proposed as a predictive biomarker to predict response to immune checkpoint blockade (ICB). We aimed to find the relationship of PD-L1 IHC to TMB and MSI using a comprehensive cancer panel assay (CCPA) with >500 genes in advanced cancer patients. CCPA results from 588 archived tissue samples were analyzed for TMB and MSI. In seven samples, whole exome sequencing confirmed TMB with Pearson’s correlation coefficient of 0.972 and all MSI-high cases were validated by pentaplex PCR. Association of TMB and MSI with their corresponding PD-L1 IHC was analyzed. The median TMB value of 588 cases was 8.25 mutations (mut)/Mb (range 0–426.8) with different distributions among the tumor types, with high proportions of high-TMB (>10mut/Mb) in tumors from melanoma, colorectal, gastric, and biliary tract. The TMB values significantly correlated with PD-L1 expression, and this correlation was prominent in gastric and biliary tract cancers. Moreover, the MSI score, the proportion of unstable MSI sites to total assessed MSI sites, showed a significant correlation with the TMB values and PD-L1 scores. This study demonstrates that PD-L1 expression is significantly associated with TMB and MSI score and this correlation depends on the location of the primary tumor.
Collapse
Affiliation(s)
- Yoon Ah Cho
- Samsung Medical Center, Department of Pathology and Translational Genomics, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (Y.A.C.); (H.L.); (H.K.); (S.Y.H.); (Y.-L.C.); (K.-T.J.)
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Korea
| | - Hyunwoo Lee
- Samsung Medical Center, Department of Pathology and Translational Genomics, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (Y.A.C.); (H.L.); (H.K.); (S.Y.H.); (Y.-L.C.); (K.-T.J.)
| | - Deok Geun Kim
- Department of Clinical Genomic Center, Samsung Medical Center, Seoul 06351, Korea;
- Department of Digital Health, Samsung Advanced Institute of Health Science and Technology, Sungkyunkwan University, Seoul 06351, Korea
| | - Hyunjin Kim
- Samsung Medical Center, Department of Pathology and Translational Genomics, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (Y.A.C.); (H.L.); (H.K.); (S.Y.H.); (Y.-L.C.); (K.-T.J.)
| | - Sang Yun Ha
- Samsung Medical Center, Department of Pathology and Translational Genomics, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (Y.A.C.); (H.L.); (H.K.); (S.Y.H.); (Y.-L.C.); (K.-T.J.)
| | - Yoon-La Choi
- Samsung Medical Center, Department of Pathology and Translational Genomics, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (Y.A.C.); (H.L.); (H.K.); (S.Y.H.); (Y.-L.C.); (K.-T.J.)
| | - Kee-Taek Jang
- Samsung Medical Center, Department of Pathology and Translational Genomics, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (Y.A.C.); (H.L.); (H.K.); (S.Y.H.); (Y.-L.C.); (K.-T.J.)
| | - Kyoung-Mee Kim
- Samsung Medical Center, Department of Pathology and Translational Genomics, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (Y.A.C.); (H.L.); (H.K.); (S.Y.H.); (Y.-L.C.); (K.-T.J.)
- Correspondence: ; Tel.: +82-2-3410-2800; Fax: +82-2-3410-6396
| |
Collapse
|
37
|
Wen Q, Yang Z, Dai H, Feng A, Li Q. Radiomics Study for Predicting the Expression of PD-L1 and Tumor Mutation Burden in Non-Small Cell Lung Cancer Based on CT Images and Clinicopathological Features. Front Oncol 2021; 11:620246. [PMID: 34422625 PMCID: PMC8377473 DOI: 10.3389/fonc.2021.620246] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Background The present study compared the predictive performance of pretreatment computed tomography (CT)-based radiomics signatures and clinicopathological and CT morphological factors for ligand programmed death-ligand 1 (PD-L1) expression level and tumor mutation burden (TMB) status and further explored predictive models in patients with advanced-stage non-small cell lung cancer (NSCLC). Methods A total of 120 patients with advanced-stage NSCLC were enrolled in this retrospective study and randomly assigned to a training dataset or validation dataset. Here, 462 radiomics features were extracted from region-of-interest (ROI) segmentation based on pretreatment CT images. The least absolute shrinkage and selection operator (LASSO) and logistic regression were applied to select radiomics features and develop combined models with clinical and morphological factors for PD-L1 expression and TMB status prediction. Ten-fold cross-validation was used to evaluate the accuracy, and the predictive performance of these models was assessed using receiver operating characteristic (ROC) and area under the curve (AUC) analyses. Results The PD-L1-positive expression level correlated with differentiation degree (p = 0.005), tumor shape (p = 0.006), and vascular convergence (p = 0.007). Stage (p = 0.023), differentiation degree (p = 0.017), and vacuole sign (p = 0.016) were associated with TMB status. Radiomics signatures showed good performance for predicting PD-L1 and TMB with AUCs of 0.730 and 0.759, respectively. Predictive models that combined radiomics signatures with clinical and morphological factors dramatically improved the predictive efficacy for PD-L1 (AUC = 0.839) and TMB (p = 0.818). The results were verified in the validation datasets. Conclusions Quantitative CT-based radiomics features have potential value in the classification of PD-L1 expression levels and TMB status. The combined model further improved the predictive performance and provided sufficient information for the guiding of immunotherapy in clinical practice, and it deserves further analysis.
Collapse
Affiliation(s)
- Qiang Wen
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhe Yang
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Honghai Dai
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Alei Feng
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiang Li
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
38
|
Liu Z, Wang T, She Y, Wu K, Gu S, Li L, Dong C, Chen C, Zhou Y. N 6-methyladenosine-modified circIGF2BP3 inhibits CD8 + T-cell responses to facilitate tumor immune evasion by promoting the deubiquitination of PD-L1 in non-small cell lung cancer. Mol Cancer 2021; 20:105. [PMID: 34416901 PMCID: PMC8377850 DOI: 10.1186/s12943-021-01398-4] [Citation(s) in RCA: 240] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/24/2021] [Indexed: 12/29/2022] Open
Abstract
Background An in-depth understanding of immune evasion mechanisms in tumors is crucial to overcome resistance and enable innovative advances in immunotherapy. Circular RNAs (circRNAs) have been implicated in cancer progression. However, much remains unknown regarding whether circRNAs impact immune escape in non-small-cell lung carcinoma (NSCLC). Methods We performed bioinformatics analysis to profile and identify the circRNAs mediating immune evasion in NSCLC. A luciferase reporter assay, RNA immunoprecipitation (RIP), RNA pulldown assays and fluorescence in situ hybridization were performed to identify the interactions among circIGF2BP3, miR-328-3p, miR-3173-5p and plakophilin 3 (PKP3). In vitro T cell-mediated killing assays and in vivo syngeneic mouse models were used to investigate the functional roles of circIGF2BP3 and its downstream target PKP3 in antitumor immunity in NSCLC. The molecular mechanism of PKP3-induced PD-L1 upregulation was explored by immunoprecipitation, RIP, and ubiquitination assays. Results We demonstrated that circIGF2BP3 (hsa_circ_0079587) expression was increased in NSCLC and negatively correlated with CD8+ T cell infiltration. Functionally, elevated circIGF2BP3 inactivated cocultured T cells in vitro and compromised antitumor immunity in an immunocompetent mouse model, and this effect was dependent on CD8+ T cells. Mechanistically, METTL3 mediates the N6-methyladenosine (m6A) modification of circIGF2BP3 and promotes its circularization in a manner dependent on the m6A reader protein YTHDC1. circIGF2BP3 competitively upregulates PKP3 expression by sponging miR-328-3p and miR-3173-5p to compromise the cancer immune response. Furthermore, PKP3 engages with the RNA-binding protein FXR1 to stabilize OTUB1 mRNA, and OTUB1 elevates PD-L1 abundance by facilitating its deubiquitination. Tumor PD-L1 deletion completely blocked the impact of the circIGF2BP3/PKP3 axis on the CD8+ T cell response. The inhibition of circIGF2BP3/PKP3 enhanced the treatment efficacy of anti-PD-1 therapy in a Lewis lung carcinoma mouse model. Collectively, the PKP3/PD-L1 signature and the infiltrating CD8+ T cell status stratified NSCLC patients into different risk groups. Conclusion Our results reveal the function of circIGF2BP3 in causing immune escape from CD8+ T cell-mediated killing through a decrease in PD-L1 ubiquitination and subsequent proteasomal degradation by stabilizing OTUB1 mRNA in a PKP3-dependent manner. This work sheds light on a novel mechanism of PD-L1 regulation in NSCLC and provides a rationale to enhance the efficacy of anti-PD-1 treatment in NSCLC. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-021-01398-4.
Collapse
Affiliation(s)
- Zhenchuan Liu
- Department of Thoracic Surgery, Shanghai Tongji Hospital, School of Medicine, Tongji University, Xincun Rd. 389, Shanghai, 200065, People's Republic of China
| | - Tingting Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Zhengmin Rd. 507, Shanghai, 200443, People's Republic of China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Zhengmin Rd. 507, Shanghai, 200443, People's Republic of China
| | - Kaiqing Wu
- Department of Thoracic Surgery, Shanghai Tongji Hospital, School of Medicine, Tongji University, Xincun Rd. 389, Shanghai, 200065, People's Republic of China
| | - Shaorui Gu
- Department of Thoracic Surgery, Shanghai Tongji Hospital, School of Medicine, Tongji University, Xincun Rd. 389, Shanghai, 200065, People's Republic of China
| | - Lei Li
- Department of Thoracic Surgery, Shanghai Tongji Hospital, School of Medicine, Tongji University, Xincun Rd. 389, Shanghai, 200065, People's Republic of China
| | - Chenglai Dong
- Department of Thoracic Surgery, Shanghai Tongji Hospital, School of Medicine, Tongji University, Xincun Rd. 389, Shanghai, 200065, People's Republic of China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Zhengmin Rd. 507, Shanghai, 200443, People's Republic of China.
| | - Yongxin Zhou
- Department of Thoracic Surgery, Shanghai Tongji Hospital, School of Medicine, Tongji University, Xincun Rd. 389, Shanghai, 200065, People's Republic of China.
| |
Collapse
|
39
|
Cheng T, Zhang J, Liu D, Lai G, Wen X. Prognosis of Non-small-cell Lung Cancer Patients With Lipid Metabolism Pathway Alternations to Immunotherapy. Front Genet 2021; 12:646362. [PMID: 34335679 PMCID: PMC8317604 DOI: 10.3389/fgene.2021.646362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) significantly improve the survival of patients with non-small-cell lung cancer (NSCLC), but only some patients obtain clinical benefits. Predictive biomarkers for ICIs can accurately identify people who will benefit from immunotherapy. Lipid metabolism signaling plays a key role in the tumor microenvironment (TME) and immunotherapy. Hence, we aimed to explore the association between the mutation status of the lipid metabolism pathway and the prognosis of patients with NSCLC treated with ICIs. We downloaded the mutation data and clinical data of a cohort of patients with NSCLC who received ICIs. Univariate and multivariate Cox regression models were used to analyze the association between the mutation status of the lipid metabolism signaling and the prognosis of NSCLC receiving ICIs. Additionally, The Cancer Genome Atlas (TCGA)–NSCLC cohort was used to explore the relationships between the different mutation statuses of lipid metabolism pathways and the TME. Additionally, we found that patients with high numbers of mutations in the lipid metabolism pathway had significantly enriched macrophages (M0- and M1-type), CD4 + T cells (activated memory), CD8 + T cells, Tfh cells and gamma delta T cells, significantly increased expression of inflammatory genes [interferon-γ (IFNG), CD8A, GZMA, GZMB, CXCL9, and CXCL10] and enhanced immunogenic factors [neoantigen loads (NALs), tumor mutation burden (TMB), and DNA damage repair pathways]. In the local-NSCLC cohort, we found that the group with a high number of mutations had a significantly higher tumor mutation burden (TMB) and PD-L1 expression. High mutation status in the lipid metabolism pathway is associated with significantly prolonged progression-free survival (PFS) in NSCLC, indicating that this marker can be used as a predictive indicator for patients with NSCLC receiving ICIs.
Collapse
Affiliation(s)
- Tianli Cheng
- Thoracic Medicine Department I, Hunan Cancer Hospital, Changsha, China.,Thoracic Medicine Department I, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | | | - Danni Liu
- HaploX Biotechnology, Shenzhen, China
| | | | - Xiaoping Wen
- Thoracic Medicine Department I, Hunan Cancer Hospital, Changsha, China.,Thoracic Medicine Department I, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| |
Collapse
|
40
|
Tong QS, Miao WM, Huang H, Luo JQ, Liu R, Huang YC, Zhao DK, Shen S, Du JZ, Wang J. A Tumor-Penetrating Nanomedicine Improves the Chemoimmunotherapy of Pancreatic Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101208. [PMID: 34145747 DOI: 10.1002/smll.202101208] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/07/2021] [Indexed: 06/12/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant tumors with a low survival rate. The therapeutic effect of chemotherapy and immunotherapy for PDAC is disappointing due to the presence of dense tumor stroma and immunosuppressive cells in the tumor microenvironment (TME). Herein, a tumor-penetrating nanoparticle is reported to modulate the deep microenvironment of PDAC for improved chemoimmunotherapy. The tumor pH-sensitive polymer is synthesized by conjugating N,N-dipentylethyl moieties and monomethoxylpoly(ethylene glycol) onto PAMAM dendrimer, into whose cavity a hydrophobic gemcitabine (Gem) prodrug is accommodated. They self-assemble into nanoparticles (denoted as SPN@Pro-Gem) with the size around 120 nm at neutral pH, but switch into small particles (≈8 nm) at tumor site to facilitate deep delivery of Gem into the tumor parenchyma. In addition to killing cancer cells that resided deeply in the tumor tissue, SPN@Pro-Gem could modulate the TME by reducing the abundance of tumor-associated macrophages and myeloid-derived suppressor cells as well as upregulating the expression level of PD-L1 of tumor cells. This collectively facilitates the infiltration of cytotoxic T cells into the tumors and renders checkpoint inhibitors more effective in previously unresponsive PDAC models. This study reveals a promising strategy for improving the chemoimmunotherapy of pancreatic cancer.
Collapse
Affiliation(s)
- Qi-Song Tong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Wei-Min Miao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Hua Huang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Jia-Qi Luo
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Rong Liu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yong-Cong Huang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Dong-Kun Zhao
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Jin-Zhi Du
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| |
Collapse
|
41
|
Jacob J, Necchi A, Grivas P, Hughes M, Sanford T, Mollapour M, Shapiro O, Talal A, Sokol E, Vergilio JA, Killian J, Lin D, Williams E, Tse J, Ramkissoon S, Severson E, Hemmerich A, Ferguson N, Edgerly C, Duncan D, Huang R, Chung J, Madison R, Alexander B, Venstrom J, Reddy P, McGregor K, Elvin J, Schrock A, Danziger N, Pavlick D, Ross J, Bratslavsky G. Comprehensive genomic profiling of histologic subtypes of urethral carcinomas. Urol Oncol 2021; 39:731.e1-731.e15. [PMID: 34215504 DOI: 10.1016/j.urolonc.2020.12.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/17/2020] [Accepted: 12/19/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND Carcinoma of the urethra (UrthCa) is an uncommon Genitourinary (GU) malignancy that can progress to advanced metastatic disease. METHODS One hundred twenty-seven metastatic UrthCa underwent hybrid capture-based comprehensive genomic profiling to evaluate all classes of genomic alterations (GA). Tumor mutational burden was determined on up to 1.1 Mbp of sequenced DNA, and microsatellite instability was determined on 114 loci. PD-L1 expression was determined by IHC (Dako 22C3). RESULTS Forty-nine (39%) urothelial (UrthUC), 31 (24%) squamous (UrthSCC), 24 (19%) adenocarcinomas NOS (UrthAC), and 12 (9%) clear cell (UrthCC) were evaluated. UrthUC and UrthSCC are more common in men; UrthAC and UrthCC are more common in women. Ages were similar in all 4 groups. GA in PIK3CA were the most frequent potentially targetable GA; mTOR pathway GA in PTEN were also identified. GA in other potentially targetable genes were also identified including ERBB2 (6% in UrthUC, 3% in UrthSCC, and 12% in UrthAC), FGFR1-3 (3% in UrthSCC), BRAF (3% in UrthAC), PTCH1 (8% in UrthCC), and MET (8% in UrthCC). Possibly reflecting their higher GA/tumor status, potential for immunotherapy benefit associated with higher tumor mutational burden and PD-L1 staining levels were seen in UrthUC and UrthSCC compared to UrthAC and UrthCC. Microsatellite instability high status was absent throughout. CONCLUSIONS Comprehensive genomic profiling reveals GA that may be predictive of both targeted and immunotherapy benefit in patients with advanced UrthCa and that could potentially be used in future adjuvant, neoadjuvant, and metastatic disease trials.
Collapse
Affiliation(s)
- Joseph Jacob
- SUNY Upstate Medical University, Department of Urology, Syracuse, NY
| | | | | | - Michael Hughes
- SUNY Upstate Medical University, Department of Urology, Syracuse, NY
| | - Thomas Sanford
- SUNY Upstate Medical University, Department of Urology, Syracuse, NY
| | - Mehdi Mollapour
- SUNY Upstate Medical University, Department of Urology, Syracuse, NY; SUNY Upstate Medical University Department of Biochemistry and Molecular Biology, Syracuse, NY
| | - Oleg Shapiro
- SUNY Upstate Medical University, Department of Urology, Syracuse, NY
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jeffrey Ross
- SUNY Upstate Medical University, Department of Urology, Syracuse, NY; Foundation Medicine, Cambridge, MA
| | - Gennady Bratslavsky
- SUNY Upstate Medical University, Department of Urology, Syracuse, NY; SUNY Upstate Medical University Department of Biochemistry and Molecular Biology, Syracuse, NY.
| |
Collapse
|
42
|
Zhang L, Chen Y, Wang H, Xu Z, Wang Y, Li S, Liu J, Chen Y, Luo H, Wu L, Yang Y, Zhang H, Peng H. Massive PD-L1 and CD8 double positive TILs characterize an immunosuppressive microenvironment with high mutational burden in lung cancer. J Immunother Cancer 2021; 9:jitc-2021-002356. [PMID: 34140315 PMCID: PMC8212410 DOI: 10.1136/jitc-2021-002356] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2021] [Indexed: 12/22/2022] Open
Abstract
Background Programmed cell death ligand 1 (PD-L1) expressed on tumor and immune cells are both associated with the response to programmed cell death 1 (PD-1) pathway blockade therapy. Here, we examine the role of CD8+PD-L1+ tumor-infiltrating lymphocyte (Tils) in the tumor microenvironment of non-small cell lung cancer (NSCLC). Methods Tumor tissue samples of a total of 378 patients from two NSCLC cohorts were collected retrospectively. Tumor genetic variations were measured by targeted next-generation sequencing of 543 oncogenes. Tils were assessed by multiplex immunohistochemistry assay. Correlations among Tils, tumor genetic variations, and clinicopathological characteristics were analyzed. Results The levels of CD8+PD-L1+ Tils varied in NSCLC tumor tissues. Tumor samples with high CD8+PD-L1+ Tils had higher levels of CD8+ Tils, CD68+ macrophages, PD-L1+ tumor cells, PD-1+ Tils, and CD163+ M2-type macrophages, and also had a higher tumor mutation burden, all of which collectively constituted a typically hot but immunosuppressive tumor microenvironment. Therefore, in a non-immunotherapy cohort, we observed that the higher the CD8+PD-L1+ Tils level in the tumor tissue, the worse the prognosis (progression-free survival; cohort A, stage I–II tumor; p=0.005). Contrarily, in an immunotherapy cohort, where the immune suppression was blocked by anti-PD-1 treatment, the higher the CD8+PD-L1+ Tils level, the better the response to the anti-PD-1 treatment (complete response/partial response vs stable disease/progressive disease; cohort B; p=0.0337). Conclusions CD8+PD-L1+ Tils may be an indicator of the hot but immunosuppressive tumor microenvironment which is related to a high tumor mutation burden. PD-1 pathway blockade therapy can help to mitigate this immunosuppression and obtain better curative effects.
Collapse
Affiliation(s)
- Libin Zhang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yanhui Chen
- Department of Medicine, Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu, China
| | - Han Wang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Zheyuan Xu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yang Wang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Sixing Li
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Jun Liu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yun Chen
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Hongli Luo
- Department of Medicine, Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu, China
| | - Lijia Wu
- Department of Bioinformatics, Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu, China
| | - Ying Yang
- Department of Bioinformatics, Genecast Biotechnology Co., Ltd, Wuxi, Jiangsu, China
| | - Henghui Zhang
- Biomedical Inovation Center, Beijing Shijitan Hospital, Capital Medical University; School of Oncology, Capital Medical University, Beijing, China
| | - Hao Peng
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| |
Collapse
|
43
|
Chen F, Song J, Ye Z, Xu B, Cheng H, Zhang S, Sun X. Integrated Analysis of Cell Cycle-Related and Immunity-Related Biomarker Signatures to Improve the Prognosis Prediction of Lung Adenocarcinoma. Front Oncol 2021; 11:666826. [PMID: 34150632 PMCID: PMC8212041 DOI: 10.3389/fonc.2021.666826] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a leading malignancy and has a poor prognosis over the decades. LUAD is characterized by dysregulation of cell cycle. Immunotherapy has emerged as an ideal option for treating LUAD. Nevertheless, optimal biomarkers to predict outcomes of immunotherapy is still ill-defined and little is known about the interaction of cell cycle-related genes (CCRGs) and immunity-related genes (IRGs). Methods We downloaded gene expression and clinical data from TCGA and GEO database. LASSO regression and Cox regression were used to construct a differentially expressed CCRGs and IRGs signature. We used Kaplan-Meier analysis to compare survival of LUAD patients. We constructed a nomogram to predict the survival and calibration curves were used to evaluate the accuracy. Results A total of 61 differentially expressed CCRGs and IRGs were screened out. We constructed a new risk model based on 8 genes, including ACVR1B, BIRC5, NR2E1, INSR, TGFA, BMP7, CD28, NUDT6. Subgroup analysis revealed the risk model accurately predicted the overall survival in LUAD patients with different clinical features and was correlated with immune cells infiltration. A nomogram based on the risk model exhibited excellent performance in survival prediction of LUAD. Conclusions The 8 gene survival signature and nomogram in our study are effective and have potential clinical application to predict prognosis of LUAD.
Collapse
Affiliation(s)
- Fangyu Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiahang Song
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ziqi Ye
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bing Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongyan Cheng
- Department of Synthetic Internal Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shu Zhang
- Core Facility Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
44
|
Galvano A, Gristina V, Malapelle U, Pisapia P, Pepe F, Barraco N, Castiglia M, Perez A, Rolfo C, Troncone G, Russo A, Bazan V. The prognostic impact of tumor mutational burden (TMB) in the first-line management of advanced non-oncogene addicted non-small-cell lung cancer (NSCLC): a systematic review and meta-analysis of randomized controlled trials. ESMO Open 2021; 6:100124. [PMID: 33940346 PMCID: PMC8111593 DOI: 10.1016/j.esmoop.2021.100124] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/30/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The role of tumor mutational burden (TMB) is still debated for selecting advanced non-oncogene addicted non-small-cell lung cancer (NSCLC) patients who might benefit from immune checkpoint inhibitors (ICIs). Of note, TMB failed to predict a benefit in overall survival (OS) among such patients. MATERIALS AND METHODS The purpose of this meta-analysis was to compare efficacy outcomes among first-line immune-oncology (IO) agents versus standard platinum-based chemotherapy (CT) within two subgroups (TMB-low and TMB-high on either tissue or blood). We collected hazard ratios (HRs) to evaluate the association for progression-free survival (PFS) and OS, with the relative 95% confidence intervals (CIs). Risk ratios (RRs) were used as an association measure for objective response rate (ORR). RESULTS Eight different cohorts of five randomized controlled phase III studies (3848 patients) were analyzed. In TMB-high patients, IO agents were associated with improved ORR (RRs 1.37, 95% CI 1.13-1.66), PFS (HR 0.69, 95% CI 0.61-0.79) and OS (HR 0.67, 95% CI 0.59-0.77) when compared with CT, thus suggesting a possible predictive role of high TMB for IO regimens. In TMB-low patients, the IO strategy did not lead to any significant benefit in survival and activity, whereas the pooled results of both ORR and PFS were intriguingly associated with a statistical significance in favor of CT. CONCLUSIONS This meta-analysis resulted in a proven benefit in OS in favor of IO agents in the TMB-high population. Although more prospective data are warranted, we postulated the hypothesis that monitoring TMB, in addition to the existing programmed death-ligand 1 (PD-L1) expression level, could represent the preferable option for future clinical research in the first-line management of advanced non-oncogene addicted NSCLC patients.
Collapse
Affiliation(s)
- A Galvano
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - V Gristina
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - U Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - P Pisapia
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - F Pepe
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - N Barraco
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - M Castiglia
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - A Perez
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - C Rolfo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA
| | - G Troncone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - A Russo
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy.
| | - V Bazan
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| |
Collapse
|
45
|
Yin L, Zhou L, Xu R. Identification of Tumor Mutation Burden and Immune Infiltrates in Hepatocellular Carcinoma Based on Multi-Omics Analysis. Front Mol Biosci 2021; 7:599142. [PMID: 33681288 PMCID: PMC7928364 DOI: 10.3389/fmolb.2020.599142] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
We aimed to explore the tumor mutational burden (TMB) and immune infiltration in HCC and investigate new biomarkers for immunotherapy. Transcriptome and gene mutation data were downloaded from the GDC portal, including 374 HCC samples and 50 matched normal samples. Furthermore, we divided the samples into high and low TMB groups, and analyzed the differential genes between them with GO, KEGG, and GSEA. Cibersort was used to assess the immune cell infiltration in the samples. Finally, univariate and multivariate Cox regression analyses were performed to identify differential genes related to TMB and immune infiltration, and a risk prediction model was constructed. We found 10 frequently mutated genes, including TP53, TTN, CTNNB1, MUC16, ALB, PCLO, MUC, APOB, RYR2, and ABCA. Pathway analysis indicated that these TMB-related differential genes were mainly enriched in PI3K-AKT. Cibersort analysis showed that memory B cells (p = 0.02), CD8+ T cells (p = 0.09), CD4+ memory activated T cells (p = 0.07), and neutrophils (p = 0.06) demonstrated a difference in immune infiltration between high and low TMB groups. On multivariate analysis, GABRA3 (p = 0.05), CECR7 (p < 0.001), TRIM16 (p = 0.003), and IL7R (p = 0.04) were associated with TMB and immune infiltration. The risk prediction model had an area under the curve (AUC) of 0.69, suggesting that patients with low risk had better survival outcomes. Our study demonstrated for the first time that CECR7, GABRA3, IL7R, and TRIM16L were associated with TMB and promoted antitumor immunity in HCC.
Collapse
Affiliation(s)
- Lu Yin
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liuzhi Zhou
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Rujun Xu
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
46
|
Qu J, Mei Q, Liu L, Cheng T, Wang P, Chen L, Zhou J. The progress and challenge of anti-PD-1/PD-L1 immunotherapy in treating non-small cell lung cancer. Ther Adv Med Oncol 2021; 13:1758835921992968. [PMID: 33643442 PMCID: PMC7890731 DOI: 10.1177/1758835921992968] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
The use of programmed cell-death protein 1 (PD-1)/programmed cell-death ligand 1 (PD-L1) inhibitors is the standard therapy for the first-line or second-line treatment of patients with non-small-cell lung cancer (NSCLC). In contrast to current traditional treatments such as chemotherapy or radiotherapy, anti-PD-1 and anti-PD-L1 treatments can directly attenuate tumour-mediated exhaustion and effectively modulate the host anti-tumour immune response in vivo. In addition, compared with traditional therapy, PD-1/PD-L1 inhibitor monotherapy can significantly prolong survival without obvious side effects in the treatment of advanced NSCLC. Ideally, several biomarkers could be used to monitor the safety and effectiveness of anti-PD-1 and anti-PD-L1 treatments; however, the current lack of optimal prognostic markers remains a widespread limitation and challenge for further clinical applications, as does the possibility of immune-related adverse events and drug resistance. In this review, we aimed to summarise the latest progress in anti-PD-1/anti-PD-L1 treatment of advanced NSCLC, worldwide, including in China. An exploration of underlying biomarker identification and future challenges will be discussed in this article to facilitate translational studies in cancer immunotherapy.
Collapse
Affiliation(s)
- Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Quanhui Mei
- Intensive Care Unit, The First People’s Hospital of Changde City, Changde, Hunan, PR China
| | - Li Liu
- Lung Cancer and Gastroenterology Department, Hunan Cancer Hospital, Affiliated Tumour Hospital of Xiangya Medical School of Central South University, Changsha, Hunan, PR China
| | - Tianli Cheng
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Affiliated Tumour Hospital of Xiangya Medical School of Central South University, Changsha, Hunan, PR China
| | - Peng Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, PR China
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, PR China
| |
Collapse
|
47
|
Amôr NG, Santos PSDS, Campanelli AP. The Tumor Microenvironment in SCC: Mechanisms and Therapeutic Opportunities. Front Cell Dev Biol 2021; 9:636544. [PMID: 33634137 PMCID: PMC7900131 DOI: 10.3389/fcell.2021.636544] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/18/2021] [Indexed: 12/14/2022] Open
Abstract
Squamous cell carcinoma (SCC) is the second most common skin cancer worldwide and, despite the relatively easy visualization of the tumor in the clinic, a sizeable number of SCC patients are diagnosed at advanced stages with local invasion and distant metastatic lesions. In the last decade, immunotherapy has emerged as the fourth pillar in cancer therapy via the targeting of immune checkpoint molecules such as programmed cell-death protein-1 (PD-1), programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). FDA-approved monoclonal antibodies directed against these immune targets have provide survival benefit in a growing list of cancer types. Currently, there are two immunotherapy drugs available for cutaneous SCC: cemiplimab and pembrolizumab; both monoclonal antibodies (mAb) that block PD-1 thereby promoting T-cell activation and/or function. However, the success rate of these checkpoint inhibitors currently remains around 50%, which means that half of the patients with advanced SCC experience no benefit from this treatment. This review will highlight the mechanisms by which the immune checkpoint molecules regulate the tumor microenvironment (TME), as well as the ongoing clinical trials that are employing single or combinatory therapeutic approaches for SCC immunotherapy. We also discuss the regulation of additional pathways that might promote superior therapeutic efficacy, and consequently provide increased survival for those patients that do not benefit from the current checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Nádia Ghinelli Amôr
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Paulo Sérgio da Silva Santos
- Department of Surgery, Stomatology, Pathology, and Radiology, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Ana Paula Campanelli
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| |
Collapse
|
48
|
Yin Z, Yu M, Ma T, Zhang C, Huang S, Karimzadeh MR, Momtazi-Borojeni AA, Chen S. Mechanisms underlying low-clinical responses to PD-1/PD-L1 blocking antibodies in immunotherapy of cancer: a key role of exosomal PD-L1. J Immunother Cancer 2021; 9:jitc-2020-001698. [PMID: 33472857 PMCID: PMC7818841 DOI: 10.1136/jitc-2020-001698] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 12/15/2022] Open
Abstract
Exosomes, as the main group of extracellular vesicles, are biologically active lipid-bilayer vesicles that are naturally released from different types of normal or tumor cells. These vesicles play an important role in intercellular communication and influence the extracellular environment and the immune system. Emerging evidence demonstrates that cancer-derived exosomes are enriched in immunosuppressive proteins, such as the programmed death-ligand 1 (PD-L1). PD-L1 and its receptor programmed cell death protein 1 (PD-1) are the key immune checkpoint molecules that promote tumor progression via negative regulation of immune responses. PDL-1 is highly expressed on the surface of tumor cells and binds to PD-1 on the surface of activated T cells, leading to suppression of T cells, which consequently enables cancer cells to escape antitumor immunity. Currently, there are several Food and Drug Administration-approved monoclonal antibodies blocking PD-1/PD-L1 interaction, which are clinically used for cancer treatment. However, despite impressive treatment outcomes, some patients show poor response to PD-1/PD-L1 blockade. Of note, tumor-derived exosomes containing PD-L1 can recapitulate the effect of cell-surface PD-L1. There is evidence that reveals a significant association between levels of circulating exosomal PD-L1 and rate of response to anti-PD-1/PD-L1 antibody therapy. The present article reviews the role of exosomal PDL-1 in the therapeutic resistance to anti-PD-1/PD-L1 treatment. Importantly, it is suggested that the removal of exosomal PDL-1 could serve as a therapeutic adjuvant for enhancing the efficacy of anti-PD-1/PD-L1 therapy in patients with cancer.
Collapse
Affiliation(s)
- Zi Yin
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Min Yu
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Tingting Ma
- Department of Obstetrics and Gynecology, Sun Yat Sen Memorial Hospital, Sun Yat sen University, Guangzhou, China
| | - Chuanzhao Zhang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Shanzhou Huang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Mohammad Reza Karimzadeh
- Department of Medical Genetics, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Amir Abaas Momtazi-Borojeni
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sheng Chen
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| |
Collapse
|
49
|
Dong A, Zhao Y, Li Z, Hu H. PD-L1 versus tumor mutation burden: Which is the better immunotherapy biomarker in advanced non-small cell lung cancer? J Gene Med 2021; 23:e3294. [PMID: 33171529 DOI: 10.1002/jgm.3294] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/24/2020] [Accepted: 10/24/2020] [Indexed: 12/31/2022] Open
Abstract
PD-L1 and tumor mutation burden (TMB) are the most widely used immunotherapy biomarkers to identify populations who would attain clinical benefit, with the higher values predicting better therapeutic efficacy. This review addresses the predictive values and unresolved challenges of these two biomarkers. PD-1 and PD-L1 inhibitors have induced durable and effective responses in patients with advanced non-small cell lung cancer, confirmed by multiple clinical trials and real-world studies. Different clinical trials, involving both PD-1/PD-L1 inhibitors alone and combination regimens, adopted either PD-L1 or TMB to stratify the patients, although the predictive capabilities of these two biomarkers are different. In the first-line setting, PD-L1 of 50% or more as a cut-off value can help select candidates for pembrolizumab or atezolizumab monotherapy; however, these two biomarkers poorly predict the efficacy of immunotherapy combination regimens as first-line treatments. In the second-line setting, although patients can benefit from nivolumab regardless of PD-L1 expression, both PD-L1 and blood TMB can be used as biomarkers to find patients suitable for atezolizumab. Except for inaccurate predictiveness, there are many unresolved problems with regard to the two biomarkers, such as the lack of standard detection methods, and their susceptibilities to other dynamic changes. The predictive values of TMB and PD-L1 were low in most circumstances; however, PD-L1 expression greater than ≥ 50% can help select appropriate patients for pembrolizumab and atezolizumab, respectively, as first-line monotherapies. Higher PD-L1 or TMB was associated with greater efficacy for atezolizumab as a second-line monotherapy.
Collapse
Affiliation(s)
- Aoran Dong
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yiming Zhao
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhihua Li
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Hai Hu
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
50
|
Yu L, Lai Q, Gou L, Feng J, Yang J. Opportunities and obstacles of targeted therapy and immunotherapy in small cell lung cancer. J Drug Target 2021; 29:1-11. [PMID: 32700566 DOI: 10.1080/1061186x.2020.1797050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 05/26/2020] [Accepted: 07/06/2020] [Indexed: 02/05/2023]
Abstract
Small cell lung cancer (SCLC) is an aggressive malignant tumour which accounts for approximately 13-15% of all newly diagnosed lung cancer cases. To date, platinum-based chemotherapy are still the first-line treatments for SCLC. However, chemotherapy resistance and systemic toxicity limit the long-term clinical outcome of first-line treatment in SCLC. Recent years, targeted therapy and immunotherapy have made great breakthrough in cancer therapy, and researchers aim to exploit both as a single agent or in combination with chemotherapy to improve the survival of SCLC patients, but limited effectiveness and the adverse events remain the major obstacles in the treatment of SCLC. To overcome these challenges for SCLC therapies, prevention and early diagnosis for this refractory disease is very important. At the same time, we should reveal more information about the pathogenesis of SCLC and the mechanism of drug resistance. Finally, new treatment strategies should also be taken into considerations, such as repurposing drug, optimising of targets, combination therapy strategies or prognostic biomarkers to enhance therapeutic effects and decrease the adverse events rates in SCLC patients. This article will review the molecular biology characteristics of SCLC and discuss the opportunities and obstacles of the current therapy for SCLC patients.
Collapse
Affiliation(s)
- Lin Yu
- The Clinical Laboratory of Mianyang Central Hospital, Mianyang, China
- Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Qinhuai Lai
- Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Lantu Gou
- Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jiafu Feng
- The Clinical Laboratory of Mianyang Central Hospital, Mianyang, China
| | - Jinliang Yang
- Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|