1
|
Su J, Li Z, Wang J, Liu N, Bao L, Du J, Li Y, Yu Y, Wang H. The causal relationship between anti-CD20 antibodies and endometrial cancer: a Mendelian randomization study. Discov Oncol 2024; 15:613. [PMID: 39487866 PMCID: PMC11531411 DOI: 10.1007/s12672-024-01491-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024] Open
Abstract
PURPOSE To determine the effects of anti-B cell surface marker CD20 antibody on the occurrence and progression of endometrial cancer to provide insights into clinical treatment in the future. METHODS We performed a two-sample Mendelian randomization analysis (MR) to analyze the causal relationship between EC and immune cell markers. The genome-wide association study (GWAS) data of MS4A1 single nucleotide polymorphism encoding gene of CD20 were collected, and a drug target MR analysis was performed to detect the causal relationship between anti-CD20 antibody and the risk of EC. The risk of follicular lymphoma was used as a positive control, and EC was used as the outcome. RESULTS EC exerted an effect on 28 immunophenotypes, predominantly on CD20. CD20 in immunoglobulin D+ CD38- B cells, memory B cells, and unsw memory B cells increased after EC onset. In addition, we detected one immunophenotype that exerted a dangerous effect on EC, i.e., CD3/TD CD4+ (mature stage of the T cell group). Simultaneously, CD20 could be used as both a risk factor and a protective factor for EC (P > 0.05). Anti-CD20 antibodies significantly reduced the risk of follicular lymphoma, including two GWAS pooled datasets. In addition, anti-CD20 antibodies exerted a protective effect on EC in two GWAS pooled datasets (P > 0.05). CONCLUSION Our study confirms the close relationship between CD20 and EC. Anti-CD20 antibodies may exert a protective effect on EC and reduce the risk of EC morbidity, providing a reference for future clinical diagnosis and treatment. However, further clinical verification of our results is warranted.
Collapse
Affiliation(s)
- Jinqiu Su
- Department of Gynecology, Affiliated Cancer Hospital of Guangxi Medical University, Guangxi Key Laboratory of Early Prevention and Treatment of Regional High-Incidence Tumors, Nanning, 530021, Guangxi, China
| | - Zhuang Li
- Department of Gynecology, Affiliated Cancer Hospital of Guangxi Medical University, Guangxi Key Laboratory of Early Prevention and Treatment of Regional High-Incidence Tumors, Nanning, 530021, Guangxi, China
| | - Jianli Wang
- Department of Gynecology, Affiliated Cancer Hospital of Guangxi Medical University, Guangxi Key Laboratory of Early Prevention and Treatment of Regional High-Incidence Tumors, Nanning, 530021, Guangxi, China
| | - Nairong Liu
- Department of Gynecology, Affiliated Cancer Hospital of Guangxi Medical University, Guangxi Key Laboratory of Early Prevention and Treatment of Regional High-Incidence Tumors, Nanning, 530021, Guangxi, China
| | - Liyi Bao
- Department of Gynecology, Affiliated Cancer Hospital of Guangxi Medical University, Guangxi Key Laboratory of Early Prevention and Treatment of Regional High-Incidence Tumors, Nanning, 530021, Guangxi, China
| | - Jiayuan Du
- Department of Gynecology, Affiliated Cancer Hospital of Guangxi Medical University, Guangxi Key Laboratory of Early Prevention and Treatment of Regional High-Incidence Tumors, Nanning, 530021, Guangxi, China
| | - Yan Li
- Department of Gynecology, Affiliated Cancer Hospital of Guangxi Medical University, Guangxi Key Laboratory of Early Prevention and Treatment of Regional High-Incidence Tumors, Nanning, 530021, Guangxi, China
| | - Yaojie Yu
- Department of Gynecology, Affiliated Cancer Hospital of Guangxi Medical University, Guangxi Key Laboratory of Early Prevention and Treatment of Regional High-Incidence Tumors, Nanning, 530021, Guangxi, China
| | - He Wang
- Department of Gynecology, Affiliated Cancer Hospital of Guangxi Medical University, Guangxi Key Laboratory of Early Prevention and Treatment of Regional High-Incidence Tumors, Nanning, 530021, Guangxi, China.
| |
Collapse
|
2
|
Lin L, Zou J, Pei S, Huang W, Zhang Y, Zhao Z, Ding Y, Xiao C. Germinal center B-cell subgroups in the tumor microenvironment cannot be overlooked: Their involvement in prognosis, immunotherapy response, and treatment resistance in head and neck squamous carcinoma. Heliyon 2024; 10:e37726. [PMID: 39391510 PMCID: PMC11466559 DOI: 10.1016/j.heliyon.2024.e37726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Background More than 60 % of patients with head and neck squamous carcinoma (HNSCC) are diagnosed at advanced stages and miss radical treatment. This has prompted the need to find new biomarkers to achieve early diagnosis and predict early recurrence and metastasis of tumors. Methods Single-cell RNA sequencing (scRNA-seq) data from HNSCC tissues and peripheral blood samples were obtained through the Gene Expression Omnibus (GEO) database (GSE164690) to characterize the B-cell subgroups, differentiation trajectories, and intercellular communication networks in HNSCC and to construct a prognostic model of the associated risks. In addition, this study analyzed the differences in clinical features, immune cell infiltration, functional enrichment, tumor mutational burden (TMB), and drug sensitivity between the high- and low-risk groups. Results Using scRNA-seq of HNSCC, we classified B and plasma cells into a total of four subgroups: naive B cells (NBs), germinal center B cells (GCBs), memory B cells (MBs), and plasma cells (PCs). Pseudotemporal trajectory analysis revealed that NBs and GCBs were at the early stage of B cell differentiation, while MBs and PCs were at the end. Cellular communication revealed that GCBs acted on tumor cells through the CD99 and SEMA4 signaling pathways. The independent prognostic value, immune cell infiltration, TMB and drug sensitivity assays were validated for the MEF2B+ GCB score groups. Conclusions We identified GCBs as B cell-specific prognostic biomarkers for the first time. The MEF2B+ GCB score fills the research gap in the genetic prognostic prediction model of HNSCC and is expected to provide a theoretical basis for finding new therapeutic targets for HNSCC.
Collapse
Affiliation(s)
- Li Lin
- Department of Stomatology, the First Affiliated Hospital of Soochow University, 188 Shi Zi Rd, Suzhou, 215006, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Jiani Zou
- China Eastern Airlines, Comprehensive Management Department, Aviation Health Department, China
| | - Shengbin Pei
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenyi Huang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Yichi Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Zhijie Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Yantao Ding
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui, 230032, China
- China bKey Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, 230032, China
| | - Can Xiao
- Department of Stomatology, the First Affiliated Hospital of Soochow University, 188 Shi Zi Rd, Suzhou, 215006, China
| |
Collapse
|
3
|
Xie M, Lin X, Bao X, Liang Y, Deng H, Song J, Ma X, Zhang X, Yao J, Pan L, Xue X. Tertiary Lymphoid Structure in Tumor Microenvironment and Immunotherapy of Lung Cancer. Arch Bronconeumol 2024; 60 Suppl 2:S77-S85. [PMID: 39174437 DOI: 10.1016/j.arbres.2024.07.020] [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: 05/08/2024] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 08/24/2024]
Abstract
Immune checkpoint inhibitors have opened an era of lung cancer therapy. However, a notable disparity exists in the efficacy of immunotherapy among individual patients. The tertiary lymphoid structure (TLS) is an ectopic lymphocyte aggregation that appears under pathological conditions and is the primary site of action for anti-tumor immunity. It is commonly reported that the presence of TLS within the tumor microenvironment (TME) relates to a favorable clinical prognosis and an excellent response to immunotherapy in lung cancer patients. A thorough understanding of TLS and its dynamic changes in TME has become an attractive focus for optimizing immunotherapy strategies for lung cancer. In this review, we comprehensively generalize the composition, formation, mechanism, detection methods of TLS, and summarize the role of TLS in lung cancer immunotherapy. Finally, induction of TLS is also discussed, which may provide more effective therapeutic strategies for lung cancer therapy.
Collapse
Affiliation(s)
- Mei Xie
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, People's Republic of China
| | - Xuwen Lin
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, People's Republic of China
| | - Xinyu Bao
- Department of Respiratory and Critical Care, Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, People's Republic of China
| | - Yiran Liang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, People's Republic of China
| | - Hui Deng
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, People's Republic of China
| | - Jialin Song
- Department of Respiratory and Critical Care, Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, People's Republic of China
| | - Xidong Ma
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, People's Republic of China
| | - Xin Zhang
- Department of Respiratory and Critical Care, Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, People's Republic of China
| | - Jie Yao
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, People's Republic of China
| | - Lei Pan
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, People's Republic of China.
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, People's Republic of China.
| |
Collapse
|
4
|
Yolmo P, Rahimi S, Chenard S, Conseil G, Jenkins D, Sachdeva K, Emon I, Hamilton J, Xu M, Rangachari M, Michaud E, Mansure JJ, Kassouf W, Berman DM, Siemens DR, Koti M. Atypical B Cells Promote Cancer Progression and Poor Response to Bacillus Calmette-Guérin in Non-Muscle Invasive Bladder Cancer. Cancer Immunol Res 2024; 12:1320-1339. [PMID: 38916567 PMCID: PMC11443217 DOI: 10.1158/2326-6066.cir-23-1114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/03/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
Poor response to Bacillus Calmette-Guérin (BCG) immunotherapy remains a major barrier in the management of patients with non-muscle invasive bladder cancer (NMIBC). Multiple factors are associated with poor outcomes, including biological aging and female sex. More recently, it has emerged that a B-cell-infiltrated pretreatment immune microenvironment of NMIBC tumors can influence the response to intravesically administered BCG. The mechanisms underlying the roles of B cells in NMIBC are poorly understood. Here, we show that B-cell-dominant tertiary lymphoid structures (TLSs), a hallmark feature of the chronic mucosal immune response, are abundant and located close to the epithelial compartment in pretreatment tumors from BCG non-responders. Digital spatial proteomic profiling of whole tumor sections from male and female patients with NMIBC who underwent treatment with intravesical BCG, revealed higher expression of immune exhaustion-associated proteins within the tumor-adjacent TLSs in both responders and non-responders. Chronic local inflammation, induced by the N-butyl-N-(4-hydroxybutyl) nitrosamine carcinogen, led to TLS formation with recruitment and differentiation of the immunosuppressive atypical B-cell (ABC) subset within the bladder microenvironment, predominantly in aging female mice compared to their male counterparts. Depletion of ABCs simultaneous to BCG treatment delayed cancer progression in female mice. Our findings provide evidence indicating a role for ABCs in BCG response and will inform future development of therapies targeting the B-cell-exhaustion axis.
Collapse
Affiliation(s)
- Priyanka Yolmo
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Sadaf Rahimi
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Stephen Chenard
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Gwenaëlle Conseil
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Danielle Jenkins
- Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Kartik Sachdeva
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Isaac Emon
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
| | - Jake Hamilton
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Minqi Xu
- Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Manu Rangachari
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Eva Michaud
- Division of Urology, Department of Surgery, McGill University Health Center, Montreal, Canada
| | - Jose J Mansure
- Division of Urology, Department of Surgery, McGill University Health Center, Montreal, Canada
| | - Wassim Kassouf
- Division of Urology, Department of Surgery, McGill University Health Center, Montreal, Canada
| | - David M Berman
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
- Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - David R Siemens
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
- Department of Urology, Queen's University, Kingston, Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Sinclair Cancer Research Institute, Queen's University, Kingston, Canada
- Department of Urology, Queen's University, Kingston, Canada
| |
Collapse
|
5
|
Lacinski RA, Dziadowicz SA, Melemai VK, Fitzpatrick B, Pisquiy JJ, Heim T, Lohse I, Schoedel KE, Llosa NJ, Weiss KR, Lindsey BA. Spatial multiplexed immunofluorescence analysis reveals coordinated cellular networks associated with overall survival in metastatic osteosarcoma. Bone Res 2024; 12:55. [PMID: 39333065 PMCID: PMC11436896 DOI: 10.1038/s41413-024-00359-z] [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: 02/05/2024] [Revised: 06/16/2024] [Accepted: 07/18/2024] [Indexed: 09/29/2024] Open
Abstract
Patients diagnosed with advanced osteosarcoma, often in the form of lung metastases, have abysmal five-year overall survival rates. The complexity of the osteosarcoma immune tumor microenvironment has been implicated in clinical trial failures of various immunotherapies. The purpose of this exploratory study was to spatially characterize the immune tumor microenvironment of metastatic osteosarcoma lung specimens. Knowledge of the coordinating cellular networks within these tissues could then lead to improved outcomes when utilizing immunotherapy for treatment of this disease. Importantly, various cell types, interactions, and cellular neighborhoods were associated with five-year survival status. Of note, increases in cellular interactions between T lymphocytes, positive for programmed cell death protein 1, and myeloid-derived suppressor cells were observed in the 5-year deceased cohort. Additionally, cellular neighborhood analysis identified an Immune-Cold Parenchyma cellular neighborhood, also associated with worse 5-year survival. Finally, the Osteosarcoma Spatial Score, which approximates effector immune activity in the immune tumor microenvironment through the spatial proximity of immune and tumor cells, was increased within 5-year survivors, suggesting improved effector signaling in this patient cohort. Ultimately, these data represent a robust spatial multiplexed immunofluorescence analysis of the metastatic osteosarcoma immune tumor microenvironment. Various communication networks, and their association with survival, were described. In the future, identification of these networks may suggest the use of specific, combinatory immunotherapeutic strategies for improved anti-tumor immune responses and outcomes in osteosarcoma.
Collapse
Affiliation(s)
- Ryan A Lacinski
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Cancer Institute, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Sebastian A Dziadowicz
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Vincent K Melemai
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Brody Fitzpatrick
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - John J Pisquiy
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Tanya Heim
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Ines Lohse
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Karen E Schoedel
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Nicolas J Llosa
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Kurt R Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Brock A Lindsey
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
6
|
Van Kerkhove O, Verfaillie S, Maes B, Cuppens K. The Adenosinergic Pathway in Non-Small Cell Lung Cancer. Cancers (Basel) 2024; 16:3142. [PMID: 39335114 PMCID: PMC11430550 DOI: 10.3390/cancers16183142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/02/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting PD-(L)1 and CTLA-4 have revolutionized the systemic treatment of non-small cell lung cancer (NSCLC), achieving impressive results. However, long-term clinical benefits are only seen in a minority of patients. Extensive research is being conducted on novel potential immune checkpoints and the mechanisms underlying ICI resistance. The tumor microenvironment (TME) plays a critical role in modulating the immune response and influencing the efficacy of ICIs. The adenosinergic pathway and extracellular adenosine (eADO) are potential targets to improve the response to ICIs in NSCLC patients. First, this review delves into the adenosinergic pathway and the impact of adenosine within the TME. Second, we provide an overview of relevant preclinical and clinical data on molecules targeting this pathway, particularly focusing on NSCLC.
Collapse
Affiliation(s)
- Olivier Van Kerkhove
- Department of Pulmonology and Thoracic Oncology and Jessa & Science, Jessa Hospital, Salvatorstraat, 3500 Hasselt, Belgium
| | - Saartje Verfaillie
- Department of Pulmonology and Thoracic Oncology and Jessa & Science, Jessa Hospital, Salvatorstraat, 3500 Hasselt, Belgium
| | - Brigitte Maes
- Laboratory for Molecular Diagnostics, Department of Laboratory Medicine, Jessa Hospital, Salvatorstraat, 3500 Hasselt, Belgium
- Faculty of Medicine and Life Sciences-LCRC, Hasselt University, 3590 Diepenbeek, Belgium
| | - Kristof Cuppens
- Department of Pulmonology and Thoracic Oncology and Jessa & Science, Jessa Hospital, Salvatorstraat, 3500 Hasselt, Belgium
- Faculty of Medicine and Life Sciences-LCRC, Hasselt University, 3590 Diepenbeek, Belgium
| |
Collapse
|
7
|
Ma L, Qin X, Yu A, Liu H, Pan D, Gao Y, Wu Z, Chen Z, Han Z. Clinicopathological and prognostic value of tertiary lymphoid structures in lung cancer: a meta-analysis. Clin Transl Oncol 2024:10.1007/s12094-024-03677-0. [PMID: 39212910 DOI: 10.1007/s12094-024-03677-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE The purpose of this meta-analysis was to examine the clinicopathological and prognostic significance of tertiary lymphocytic infiltrates in lung cancer. METHOD A systematic search was performed in many databases, including PubMed, Web of Science, Cochrane Library, Embase, CNKI, Wangfangdate, and CBM, up until January 2024. We calculated the hazard ratio (HR), odds ratios (OR), and confidence interval (CI), and accomplished this meta-analysis with Stata 15 software. RESULT 14 studies, including 3101 patients, were subjected to analysis. High TLS detection was associated with a longer OS (HR = 0.545, 95% CI: 0.359-0.827, p = 0.004), DFS (HR = 0.431, 95% CI: 0.350-0.531, p < 0.001), and RFS (HR = 0.430, 95% CI: 0.325-0.569, p < 0.001). Meanwhile, it was observed that a higher detection of TLS was significantly correlated with the administration of adjuvant chemotherapy (OR = 1.505, 95% CI: 1.017-2.225, p = 0.041). Not only that, but there was a higher occurrence of significantly elevated TLS detection in the early N stages (N = 0) compared to the advanced N stages (N = 1, 2, and 3) (OR = 1.604, 95% CI: 1.021-2.521, p = 0.04). CONCLUSION Elevated detection of TLS has been observed to be correlated with extended OS, DFS, and RFS in cases of lung cancer. This finding suggests that TLS could potentially serve as a valuable prognostic biomarker for lung cancer.
Collapse
Affiliation(s)
- Luyao Ma
- The Affiliated Hospital of Xuzhou Medical University, Quanshan District, No. 99 West Huaihai Road, Xuzhou, Jiangsu, China
| | - Xiaobing Qin
- The Affiliated Hospital of Xuzhou Medical University, Quanshan District, No. 99 West Huaihai Road, Xuzhou, Jiangsu, China
| | - Aoyang Yu
- The Affiliated Hospital of Xuzhou Medical University, Quanshan District, No. 99 West Huaihai Road, Xuzhou, Jiangsu, China
| | - Haonan Liu
- The Affiliated Hospital of Xuzhou Medical University, Quanshan District, No. 99 West Huaihai Road, Xuzhou, Jiangsu, China
| | - Di Pan
- The Affiliated Hospital of Xuzhou Medical University, Quanshan District, No. 99 West Huaihai Road, Xuzhou, Jiangsu, China
| | - Ying Gao
- The Affiliated Hospital of Xuzhou Medical University, Quanshan District, No. 99 West Huaihai Road, Xuzhou, Jiangsu, China
| | - Zichen Wu
- The Affiliated Hospital of Xuzhou Medical University, Quanshan District, No. 99 West Huaihai Road, Xuzhou, Jiangsu, China
| | - Zihan Chen
- The Affiliated Hospital of Xuzhou Medical University, Quanshan District, No. 99 West Huaihai Road, Xuzhou, Jiangsu, China
| | - Zhengxiang Han
- The Affiliated Hospital of Xuzhou Medical University, Quanshan District, No. 99 West Huaihai Road, Xuzhou, Jiangsu, China.
| |
Collapse
|
8
|
Yao M, Yang T, Li Q, Zhang X, Zheng Z, Li J, Huang J, Sun Y, Gao X, Fang C. Characteristics and clinical significance of tertiary lymphoid structures in OSCC. Oral Dis 2024. [PMID: 39049571 DOI: 10.1111/odi.15080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/13/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVES This study aimed to investigate the characteristics of tertiary lymphoid structures (TLSs) in oral squamous cell carcinoma (OSCC) and their association with clinical and pathological features. MATERIALS AND METHODS 12 TLS-related chemokines in TCGA database were analyzed to investigate the TLSs in OSCC. The density, maturity, and location of TLSs in a large cohort of 189 OSCC patients (114 of which had clinical and prognostic information) were assessed. And the significance between TLSs and clinicopathologic characteristics was analyzed. RESULTS Bioinformatics and analysis showed that TLSs were associated with better clinical outcomes in OSCC. Histological staining and analysis showed that the overall survival rate of the high-density group (71/112, 63.4%) was significantly higher (p < 0.0001) than that of the low-density group (41/112, 36.6%), and the high-density group had fewer lymph node metastases (50.0%/68.3%, p = 0.021). And TLSs were divided into 4 types according to the maturity and location. Different types of TLSs are associated with prognosis (OS, p < 0.0001), clinical features (T stage, p = 0.028; degree of differentiation, p = 0.043), and precancerous lesion types (OSF, p = 0.049) of OSCC patients. CONCLUSION TLSs were closely associated with better OSCC prognosis, and a more systematic classification may better guide the formulation of further treatment options.
Collapse
Affiliation(s)
- Mianfeng Yao
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Tianru Yang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Qiulan Li
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Xinle Zhang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Ziran Zheng
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Jiang Li
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Jiajun Huang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Yuanxin Sun
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Xing Gao
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Changyun Fang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| |
Collapse
|
9
|
Ma L, Li R, Liu X, Yu W, Tang Z, Shen Y, Tian H. Prognostic and clinicopathological significance of tertiary lymphoid structure in non-small cell lung cancer: a systematic review and meta-analysis. BMC Cancer 2024; 24:815. [PMID: 38977962 PMCID: PMC11229181 DOI: 10.1186/s12885-024-12587-x] [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: 04/22/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the primary reason for cancer-related deaths globally. Tertiary lymphoid structure (TLS) is an organized collection of immune cells acquired in non-physiological, non-lymphoid tissues. High expression of TLS in tumor tissues is generally associated with better prognosis. This research aimed to investigate the prognostic and clinicopathological significance of TLS in patients with NSCLC. METHODS A comprehensive literature search was conducted based on Pubmed, EMBASE, and Cochrane Library databases to identify eligible studies published up to December 8, 2023. The prognostic significance and clinicopathological value of TLS in NSCLC were evaluated by calculating the combined hazard ratios (HRs) and odds ratios (ORs) and their 95% confidence intervals (CIs). Following that, additional analyses, including subgroup analysis and sensitivity analysis, were conducted. RESULTS This meta-analysis evaluated the prognostic and clinicopathological significance of TLS in 10 studies involving 1,451 patients with NSCLC. The results revealed that the high levels of TLS were strongly associated with better overall survival (OS) (HR = 0.48, 95% CI: 0.35-0.66, p < 0.001), disease-free survival (DFS)/recurrence-free survival (RFS) (HR = 0.37, 95% CI: 0.24-0.54, p < 0.001), and disease-specific survival (DSS) (HR = 0.45, 95% CI: 0.30-0.68, p < 0.001) in NSCLC patients. In addition, the increased expression of TLS was closely related to the Tumor Node Metastasis (TNM) stage of tumors (OR = 0.71, 95% CI: 0.51-1.00, p < 0.05) and neutrophil-lymphocyte ratio (NLR) (OR = 0.33, 95% CI: 0.17-0.62, p < 0.001). CONCLUSIONS The results revealed that highly expressed TLS is closely associated with a better prognosis in NSCLC patients. TLS may serve as a novel biomarker to predict the prognosis of NSCLC patients and guide the clinical treatment decisions.
Collapse
Affiliation(s)
- Luyuan Ma
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Rongyang Li
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xiaomeng Liu
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Wenhao Yu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Zhanpeng Tang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yi Shen
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
10
|
Merali N, Jessel MD, Arbe-Barnes EH, Ruby Lee WY, Gismondi M, Chouari T, O'Brien JW, Patel B, Osei-Bordom D, Rockall TA, Sivakumar S, Annels N, Frampton AE. Impact of tertiary lymphoid structures on prognosis and therapeutic response in pancreatic ductal adenocarcinoma. HPB (Oxford) 2024; 26:873-894. [PMID: 38729813 DOI: 10.1016/j.hpb.2024.04.009] [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] [Received: 01/25/2024] [Revised: 03/27/2024] [Accepted: 04/19/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is known to have a heterogeneous desmoplastic tumour microenvironment (TME) with a large number of immunosuppressive cells. Recently, high B-cell infiltration in PDAC has received growing interest as a potential therapeutic target. METHODS Our literature review summarises the characteristics of tumour-associated tertiary lymphoid structures (TLSs) and highlight the key studies exploring the clinical outcomes of TLSs in PDAC patients and the direct effect on the TME. RESULTS The location, density and maturity stages of TLSs within tumours play a key role in determining the prognosis and is a new emerging target in cancer immunotherapy. DISCUSSION TLS development is imperative to improve the prognosis of PDAC patients. In the future, studying the genetics and immune characteristics of tumour infiltrating B cells and TLSs may lead towards enhancing adaptive immunity in PDAC and designing personalised therapies.
Collapse
Affiliation(s)
- Nabeel Merali
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK; Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK; Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - Maria-Danae Jessel
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK
| | - Edward H Arbe-Barnes
- UCL Institute of Immunity and Transplantation, The Pears Building, Pond Street, London, UK
| | - Wing Yu Ruby Lee
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Martha Gismondi
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - Tarak Chouari
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK; Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - James W O'Brien
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - Bhavik Patel
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK; Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK; Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - Daniel Osei-Bordom
- Liver and Digestive Health, University College London, Royal Free Hospital, Pond St, London, UK
| | - Timothy A Rockall
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK
| | - Shivan Sivakumar
- Oncology Department and Institute of Immunology and Immunotherapy, Birmingham Medical School, University of Birmingham, Birmingham, UK
| | - Nicola Annels
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK
| | - Adam E Frampton
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, UK; Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK; Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, UK.
| |
Collapse
|
11
|
Zhang Y, Xu M, Ren Y, Ba Y, Liu S, Zuo A, Xu H, Weng S, Han X, Liu Z. Tertiary lymphoid structural heterogeneity determines tumour immunity and prospects for clinical application. Mol Cancer 2024; 23:75. [PMID: 38582847 PMCID: PMC10998345 DOI: 10.1186/s12943-024-01980-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/05/2024] [Indexed: 04/08/2024] Open
Abstract
Tertiary lymphoid structures (TLS) are clusters of immune cells that resemble and function similarly to secondary lymphoid organs (SLOs). While TLS is generally associated with an anti-tumour immune response in most cancer types, it has also been observed to act as a pro-tumour immune response. The heterogeneity of TLS function is largely determined by the composition of tumour-infiltrating lymphocytes (TILs) and the balance of cell subsets within the tumour-associated TLS (TA-TLS). TA-TLS of varying maturity, density, and location may have opposing effects on tumour immunity. Higher maturity and/or higher density TLS are often associated with favorable clinical outcomes and immunotherapeutic response, mainly due to crosstalk between different proportions of immune cell subpopulations in TA-TLS. Therefore, TLS can be used as a marker to predict the efficacy of immunotherapy in immune checkpoint blockade (ICB). Developing efficient imaging and induction methods to study TA-TLS is crucial for enhancing anti-tumour immunity. The integration of imaging techniques with biological materials, including nanoprobes and hydrogels, alongside artificial intelligence (AI), enables non-invasive in vivo visualization of TLS. In this review, we explore the dynamic interactions among T and B cell subpopulations of varying phenotypes that contribute to the structural and functional diversity of TLS, examining both existing and emerging techniques for TLS imaging and induction, focusing on cancer immunotherapies and biomaterials. We also highlight novel therapeutic approaches of TLS that are being explored with the aim of increasing ICB treatment efficacy and predicting prognosis.
Collapse
Affiliation(s)
- Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Mengjun Xu
- Medical School of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
12
|
Xie M, Gao J, Ma X, Song J, Wu C, Zhou Y, Jiang T, Liang Y, Yang C, Bao X, Zhang X, Yao J, Jing Y, Wu J, Wang J, Xue X. The radiological characteristics, tertiary lymphoid structures, and survival status associated with EGFR mutation in patients with subsolid nodules like stage I-II LUAD. BMC Cancer 2024; 24:372. [PMID: 38528507 DOI: 10.1186/s12885-024-12136-6] [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: 11/09/2023] [Accepted: 03/17/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) recommended for the patients with subsolid nodule in early lung cancer stage is not routinely. The clinical value and impact in patients with EGFR mutation on survival outcomes is further needed to be elucidated to decide whether the application of EGFR-TKIs was appropriate in early lung adenocarcinoma (LUAD) stage appearing as subsolid nodules. MATERIALS AND METHODS The inclusion of patients exhibiting clinical staging of IA-IIB subsolid nodules. Clinical information, computed tomography (CT) features before surgical resection and pathological characteristics including tertiary lymphoid structures of the tumors were recorded for further exploration of correlation with EGFR mutation and prognosis. RESULTS Finally, 325 patients were enrolled into this study, with an average age of 56.8 ± 9.8 years. There are 173 patients (53.2%) harboring EGFR mutation. Logistic regression model analysis showed that female (OR = 1.944, p = 0.015), mix ground glass nodule (OR = 2.071, p = 0.003, bubble-like lucency (OR = 1.991, p = 0.003) were significant risk factors of EGFR mutations. Additionally, EGFR mutations were negatively correlated with TLS presence and density. Prognosis analysis showed that the presence of TLS was associated with better recurrence-free survival (RFS)(p = 0.03) while EGFR mutations were associated with worse RFS(p = 0.01). The RFS in patients with TLS was considerably excel those without TLS within EGFR wild type group(p = 0.018). Multivariate analyses confirmed that EGFR mutation was an independent prognostic predictor for RFS (HR = 3.205, p = 0.037). CONCLUSIONS In early-phase LUADs, subsolid nodules with EGFR mutation had specific clinical and radiological signatures. EGFR mutation was associated with worse survival outcomes and negatively correlated with TLS, which might weaken the positive impact of TLS on prognosis. Highly attention should be paid to the use of EGFR-TKI for further treatment as agents in early LUAD patients who carrying EGFR mutation.
Collapse
Affiliation(s)
- Mei Xie
- Department of Respiratory and Critical Care, Chinese PLA General Hospital, the First Medical Centre, 100835, Beijing, People's Republic of China
| | - Jie Gao
- Department of Pathology, Chinese PLA General Hospital, the First Medical Centre, 100835, Beijing, People's Republic of China
| | - Xidong Ma
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, 100038, Beijing, People's Republic of China
| | - Jialin Song
- Department of Respiratory and Critical Care, Weifang Medical College, 261053, Weifang, People's Republic of China
| | - Chongchong Wu
- Department of Radiology, Chinese PLA General Hospital, the First Medical Centre, 100835, Beijing, People's Republic of China
| | - Yangyu Zhou
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, 100038, Beijing, People's Republic of China
| | - Tianjiao Jiang
- Department of Radiology, Affiliated Hospital of Qingdao University, 266500, Qingdao, People's Republic of China
| | - Yiran Liang
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, 100038, Beijing, People's Republic of China
| | - Chen Yang
- Department of Laboratory Medicine, Chinese PLA General Hospital, the First Medical Centre, 100835, Beijing, People's Republic of China
| | - Xinyu Bao
- Department of Respiratory and Critical Care, Weifang Medical College, 261053, Weifang, People's Republic of China
| | - Xin Zhang
- Department of Respiratory and Critical Care, Weifang Medical College, 261053, Weifang, People's Republic of China
| | - Jie Yao
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, 100038, Beijing, People's Republic of China
| | - Ying Jing
- Center for Intelligent Medicine, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, 510000, Guangzhou, People's Republic of China.
| | - Jianlin Wu
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, 116001, Dalian, People's Republic of China.
| | - Jianxin Wang
- Department of Respiratory and Critical Care, Chinese PLA General Hospital, the First Medical Centre, 100835, Beijing, People's Republic of China.
| | - Xinying Xue
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, 100038, Beijing, People's Republic of China.
| |
Collapse
|
13
|
Bryushkova EA, Mushenkova NV, Turchaninova MA, Lukyanov DK, Chudakov DM, Serebrovskaya EO. B cell clonality in cancer. Semin Immunol 2024; 72:101874. [PMID: 38508089 DOI: 10.1016/j.smim.2024.101874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 03/22/2024]
Abstract
Carcinogenesis in the process of long-term co-evolution of tumor cells and immune environment essentially becomes possible due to incorrect decisions made, remembered, and reproduced by the immune system at the level of clonal populations of antigen-specific T- and B-lymphocytes. Tumor-immunity interaction determines the nature of such errors and, consequently, delineates the possible ways of successful immunotherapeutic intervention. It is generally recognized that tumor-infiltrating B cells (TIL-B) can play both pro-tumor and anti-tumor roles. However, the exact mechanisms that determine the contribution of clonal B cell lineages with different specificities and functions remain largely unclear. This is due to the variability of cancer types, the molecular heterogeneity of tumor cells, and, to a large extent, the individual pattern of each immune response. Further progress requires detailed investigation of the functional properties and phenotypes of clonally heterogeneous B cells in relation to their antigenic specificities, which determine the functionality of both effector B lymphocytes and immunoglobulins produced in the tumor environment. Based on a real understanding of the role of clonal antigen-specific populations of B lymphocytes in the tumor microenvironment, we need to learn how to develop new methods of targeted immunotherapy, as well as adapt existing treatment options to the specific needs of different patients and patient subgroups. In this review, we will cover B cells functional diversity and their multifaceted roles in the tumor environment.
Collapse
Affiliation(s)
- E A Bryushkova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia; Department of Molecular Biology, Lomonosov Moscow State University, Moscow, Russia
| | - N V Mushenkova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia; Unicorn Capital Partners, Moscow, Russia
| | - M A Turchaninova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - D K Lukyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia; Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - D M Chudakov
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia; Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia; Central European Institute of Technology, Masaryk University, Brno, Czech Republic.
| | - E O Serebrovskaya
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia; Current position: Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| |
Collapse
|
14
|
Yuan H, Mao X, Yan Y, Huang R, Zhang Q, Zeng Y, Bao M, Dai Y, Fang B, Mi J, Xie Y, Wang X, Zhang H, Mo Z, Yang R. Single-cell sequencing reveals the heterogeneity of B cells and tertiary lymphoid structures in muscle-invasive bladder cancer. J Transl Med 2024; 22:48. [PMID: 38216927 PMCID: PMC10787393 DOI: 10.1186/s12967-024-04860-1] [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: 11/20/2023] [Accepted: 01/04/2024] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Muscle-invasive bladder cancer (MIBC) is a highly aggressive disease with a poor prognosis. B cells are crucial factors in tumor suppression, and tertiary lymphoid structures (TLSs) facilitate immune cell recruitment to the tumor microenvironment (TME). However, the function and mechanisms of tumor-infiltrating B cells and TLSs in MIBC need to be explored further. METHODS We performed single-cell RNA sequencing analysis of 11,612 B cells and 55,392 T cells from 12 bladder cancer patients and found naïve B cells, proliferating B cells, plasma cells, interferon-stimulated B cells and germinal center-associated B cells, and described the phenotype, gene enrichment, cell-cell communication, biological processes. We utilized immunohistochemistry (IHC) and immunofluorescence (IF) to describe TLSs morphology in MIBC. RESULTS The interferon-stimulated B-cell subtype (B-ISG15) and germinal center-associated B-cell subtypes (B-LMO2, B-STMN1) were significantly enriched in MIBC. TLSs in MIBC exhibited a distinct follicular structure characterized by a central region of B cells resembling a germinal center surrounded by T cells. CellChat analysis showed that CXCL13 + T cells play a pivotal role in recruiting CXCR5 + B cells. Cell migration experiments demonstrated the chemoattraction of CXCL13 toward CXCR5 + B cells. Importantly, the infiltration of the interferon-stimulated B-cell subtype and the presence of TLSs correlated with a more favorable prognosis in MIBC. CONCLUSIONS The study revealed the heterogeneity of B-cell subtypes in MIBC and suggests a pivotal role of TLSs in MIBC outcomes. Our study provides novel insights that contribute to the precision treatment of MIBC.
Collapse
Affiliation(s)
- Hao Yuan
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xingning Mao
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed By the Province and Ministry, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yunkun Yan
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Rong Huang
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Qingyun Zhang
- Department of Urology, the Affiliated Tumor Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yanyu Zeng
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Mengying Bao
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yan Dai
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Bo Fang
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Junhao Mi
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yuli Xie
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiang Wang
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Haiying Zhang
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed By the Province and Ministry, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Rirong Yang
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
15
|
Shirasawa M, Yoshida T, Ohe Y. Biomarkers of immunotherapy for non-small cell lung cancer. Jpn J Clin Oncol 2024; 54:13-22. [PMID: 37823218 DOI: 10.1093/jjco/hyad134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 09/22/2023] [Indexed: 10/13/2023] Open
Abstract
Immunotherapy is revolutionizing the treatment of non-small cell lung cancer by targeting immune checkpoint proteins, including programmed death-1, programmed death ligand 1 and cytotoxic T-lymphocyte-associated antigen 4. Several immune checkpoint inhibitors, including programmed death ligand 1 inhibitors, programmed death-1 inhibitors and cytotoxic T-lymphocyte-associated antigen 4 inhibitors, were approved for the treatment of patients with advanced non-small cell lung cancer. Programmed death ligand 1 expression is currently the only predictive biomarker for immune checkpoint inhibitors to guide the treatment strategy in these patients. However, programmed death ligand 1 expression is not a perfect biomarker for predicting the efficacy of immunotherapy. Therefore, various biomarkers such as tumour mutation burden, tumour microenvironment, gut microbiome and T-cell receptor repertoire have been proposed to predict the efficacy of immunotherapy more accurately. Additionally, combining different biomarkers may provide a more accurate prediction of response to immunotherapy. This article reports the review of the latest evidence of the predictive marker of immunotherapy in patients with advanced non-small cell lung cancer.
Collapse
Affiliation(s)
- Masayuki Shirasawa
- Department of Thoracic Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo 104-0045 Japan
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara City, Kanagawa 252-0375, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo 104-0045 Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo 104-0045 Japan
| |
Collapse
|
16
|
Tang R, Wang H, Tang M. Roles of tissue-resident immune cells in immunotherapy of non-small cell lung cancer. Front Immunol 2023; 14:1332814. [PMID: 38130725 PMCID: PMC10733439 DOI: 10.3389/fimmu.2023.1332814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common and lethal type of lung cancer, with limited treatment options and poor prognosis. Immunotherapy offers hope for improving the survival and quality of life of NSCLC patients, but its efficacy depends on the tumor immune microenvironment (TME). Tissue-resident immune cells are a subset of immune cells that reside in various tissues and organs, and play an important role in fighting tumors. In NSCLC, tissue-resident immune cells are heterogeneous in their distribution, phenotype, and function, and can either promote or inhibit tumor progression and response to immunotherapy. In this review, we summarize the current understanding on the characteristics, interactions, and roles of tissue-resident immune cells in NSCLC. We also discuss the potential applications of tissue-resident immune cells in NSCLC immunotherapy, including immune checkpoint inhibitors (ICIs), other immunomodulatory agents, and personalized cell-based therapies. We highlight the challenges and opportunities for developing targeted therapies for tissue-resident immune cells and optimizing existing immunotherapeutic approaches for NSCLC patients. We propose that tissue-resident immune cells are a key determinant of NSCLC outcome and immunotherapy response, and warrant further investigation in future research.
Collapse
Affiliation(s)
- Rui Tang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Haitao Wang
- The School of Clinical Medical Sciences, Southwest Medical University, Sichuan, Luzhou, China
| | - Mingxi Tang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Yaan People’s Hospital (Yaan Hospital of West China Hospital of Sichuan University), Yaan, Sichuan, China
| |
Collapse
|
17
|
You X, Koop K, Weigert A. Heterogeneity of tertiary lymphoid structures in cancer. Front Immunol 2023; 14:1286850. [PMID: 38111571 PMCID: PMC10725932 DOI: 10.3389/fimmu.2023.1286850] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023] Open
Abstract
The success of immunotherapy approaches, such as immune checkpoint blockade and cellular immunotherapy with genetically modified lymphocytes, has firmly embedded the immune system in the roadmap for combating cancer. Unfortunately, the majority of cancer patients do not yet benefit from these therapeutic approaches, even when the prognostic relevance of the immune response in their tumor entity has been demonstrated. Therefore, there is a justified need to explore new strategies for inducing anti-tumor immunity. The recent connection between the formation of ectopic lymphoid aggregates at tumor sites and patient prognosis, along with an effective anti-tumor response, suggests that manipulating the occurrence of these tertiary lymphoid structures (TLS) may play a critical role in activating the immune system against a growing tumor. However, mechanisms governing TLS formation and a clear understanding of their substantial heterogeneity are still lacking. Here, we briefly summarize the current state of knowledge regarding the mechanisms driving TLS development, outline the impact of TLS heterogeneity on clinical outcomes in cancer patients, and discuss appropriate systems for modeling TLS heterogeneity that may help identify new strategies for inducing protective TLS formation in cancer patients.
Collapse
Affiliation(s)
- Xin You
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Frankfurt, Germany
| | - Kristina Koop
- First Department of Medicine, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Weigert
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
- Cardiopulmonary Institute (CPI), Frankfurt, Germany
| |
Collapse
|
18
|
Yang M, Che Y, Li K, Fang Z, Li S, Wang M, Zhang Y, Xu Z, Luo L, Wu C, Lai X, Wang W. Detection and quantitative analysis of tumor-associated tertiary lymphoid structures. J Zhejiang Univ Sci B 2023; 24:779-795. [PMID: 37701955 PMCID: PMC10500099 DOI: 10.1631/jzus.b2200605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/27/2023] [Indexed: 09/14/2023]
Abstract
Tumor-associated tertiary lymphoid structures (TLSs) are ectopic lymphoid formations within tumor tissue, with mainly B and T cell populations forming the organic aggregates. The presence of TLSs in tumors has been strongly associated with patient responsiveness to immunotherapy regimens and improving tumor prognosis. Researchers have been motivated to actively explore TLSs due to their bright clinical application prospects. Various studies have attempted to decipher TLSs regarding their formation mechanism, structural composition, induction generation, predictive markers, and clinical utilization. Meanwhile, the scientific approaches to qualitative and quantitative descriptions are crucial for TLS studies. In terms of detection, hematoxylin and eosin (H&E), multiplex immunohistochemistry (mIHC), multiplex immunofluorescence (mIF), and 12-chemokine gene signature have been the top approved methods. However, no standard methods exist for the quantitative analysis of TLSs, such as absolute TLS count, analysis of TLS constituent cells, structural features, TLS spatial location, density, and maturity. This study reviews the latest research progress on TLS detection and quantification, proposes new directions for TLS assessment, and addresses issues for the quantitative application of TLSs in the clinic.
Collapse
Affiliation(s)
- Man Yang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Yurou Che
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Kezhen Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- Department of Oncology, School of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Zengyi Fang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Simin Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- Department of Oncology, School of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Mei Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Yiyao Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Zhu Xu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- Department of Oncology, School of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Liping Luo
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Chuan Wu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Xin Lai
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Weidong Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China.
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China.
- Department of Oncology, School of Clinical Medicine, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
19
|
Seethapathy H, Mistry K, Sise ME. Immunological mechanisms underlying clinical phenotypes and noninvasive diagnosis of immune checkpoint inhibitor-induced kidney disease. Immunol Rev 2023; 318:61-69. [PMID: 37482912 PMCID: PMC10865966 DOI: 10.1111/imr.13243] [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: 03/23/2023] [Accepted: 06/28/2023] [Indexed: 07/25/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have become a mainstay of cancer therapy, with over 80 FDA-approved indications. Used in a variety of settings and in combination with each other and with traditional chemotherapies, the hyperactive immune response induced by ICIs can often lead to immune-related adverse events in bystander normal tissues such as the kidneys, lungs, and the heart. In the kidneys, this immune-related adverse event manifests as acute interstitial nephritis (ICI-AIN). In the era of widespread ICI use, it becomes vital to understand the clinical manifestations of ICI-AIN and the importance of prompt diagnosis and management of these complications. In this review, we delve into the clinical phenotypes of ICI-AIN and how they differ from traditional drug-induced AIN. We also detail what is known about the mechanistic underpinnings of ICI-AIN and the important diagnostic and therapeutic implications behind harnessing those mechanisms to further our understanding of these events and to formulate effective treatment plans to manage ICI-AIN.
Collapse
Affiliation(s)
- Harish Seethapathy
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kavita Mistry
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Meghan E. Sise
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Zhou C, Qin Y, Zhao W, Liang Z, Li M, Liu D, Bai L, Chen Y, Chen Y, Cheng Y, Chu T, Chu Q, Deng H, Dong Y, Fang W, Fu X, Gao B, Han Y, He Y, Hong Q, Hu J, Hu Y, Jiang L, Jin Y, Lan F, Li Q, Li S, Li W, Li Y, Liang W, Lin G, Lin X, Liu M, Liu X, Liu X, Liu Z, Lv T, Mu C, Ouyang M, Qin J, Ren S, Shi H, Shi M, Su C, Su J, Sun D, Sun Y, Tang H, Wang H, Wang K, Wang K, Wang M, Wang Q, Wang W, Wang X, Wang Y, Wang Z, Wang Z, Wu L, Wu D, Xie B, Xie M, Xie X, Xie Z, Xu S, Xu X, Yang X, Yin Y, Yu Z, Zhang J, Zhang J, Zhang J, Zhang X, Zhang Y, Zhong D, Zhou Q, Zhou X, Zhou Y, Zhu B, Zhu Z, Zou C, Zhong N, He J, Bai C, Hu C, Li W, Song Y, Zhou J, Han B, Varga J, Barreiro E, Park HY, Petrella F, Saito Y, Goto T, Igai H, Bravaccini S, Zanoni M, Solli P, Watanabe S, Fiorelli A, Nakada T, Ichiki Y, Berardi R, Tsoukalas N, Girard N, Rossi A, Passaro A, Hida T, Li S, Chen L, Chen R. International expert consensus on diagnosis and treatment of lung cancer complicated by chronic obstructive pulmonary disease. Transl Lung Cancer Res 2023; 12:1661-1701. [PMID: 37691866 PMCID: PMC10483081 DOI: 10.21037/tlcr-23-339] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2023]
Abstract
Background Lung cancer combined by chronic obstructive pulmonary disease (LC-COPD) is a common comorbidity and their interaction with each other poses significant clinical challenges. However, there is a lack of well-established consensus on the diagnosis and treatment of LC-COPD. Methods A panel of experts, comprising specialists in oncology, respiratory medicine, radiology, interventional medicine, and thoracic surgery, was convened. The panel was presented with a comprehensive review of the current evidence pertaining to LC-COPD. After thorough discussions, the panel reached a consensus on 17 recommendations with over 70% agreement in voting to enhance the management of LC-COPD and optimize the care of these patients. Results The 17 statements focused on pathogenic mechanisms (n=2), general strategies (n=4), and clinical application in COPD (n=2) and lung cancer (n=9) were developed and modified. These statements provide guidance on early screening and treatment selection of LC-COPD, the interplay of lung cancer and COPD on treatment, and considerations during treatment. This consensus also emphasizes patient-centered and personalized treatment in the management of LC-COPD. Conclusions The consensus highlights the need for concurrent treatment for both lung cancer and COPD in LC-COPD patients, while being mindful of the mutual influence of the two conditions on treatment and monitoring for adverse reactions.
Collapse
Affiliation(s)
- Chengzhi Zhou
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Yinyin Qin
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Wei Zhao
- Department of Respiratory and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhenyu Liang
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Min Li
- Department of Respiratory Medicine, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
| | - Dan Liu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Li Bai
- Department of Respiratory Medicine, Xinqiao Hospital Army Medical University, Chongqing, China
| | - Yahong Chen
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yan Chen
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Cheng
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Tianqing Chu
- Department of Respiratory Medicine, Shanghai Chest Hospital, Jiaotong University, Shanghai, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Haiyi Deng
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Yuchao Dong
- Department of Pulmonary and Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wenfeng Fang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiuhua Fu
- Division of Respiratory Diseases, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Beili Gao
- Department of Respiratory, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiping Han
- Department of Respiratory Medicine, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yong He
- Department of Pulmonary and Critical Care Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Qunying Hong
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Hu
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Hu
- Department of Medical Oncology, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Liyan Jiang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Jiaotong University, Shanghai, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fen Lan
- Department of Respiratory Medicine, The Second Affiliated Hospital of Zhejiang University of Medicine, Hangzhou, China
| | - Qiang Li
- Department of Respiratory Medicine, Shanghai Dongfang Hospital, Shanghai, China
| | - Shuben Li
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yaqing Li
- Department of Internal Medicine, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Wenhua Liang
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Gen Lin
- Department of Thoracic Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Xinqing Lin
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Ming Liu
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Xiaofang Liu
- Department of Respiratory and Critical Care Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaoju Liu
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhefeng Liu
- Department of Oncology, General Hospital of Chinese PLA, Beijing, China
| | - Tangfeng Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Chuanyong Mu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming Ouyang
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Jianwen Qin
- Department of Respiratory and Critical Care Medicine, Tianjin Chest Hospital, Tianjin, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Huanzhong Shi
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Minhua Shi
- Department of Respiratory Medicine, The Second Affiliated Hospital of Suzhou University, Suzhou, China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin Su
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dejun Sun
- Department of Respiratory and Critical Care Medicine, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, China
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Huaping Tang
- Department of Respiratory Medicine, Qingdao Municipal Hospital, Qingdao, China
| | - Huijuan Wang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Kai Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Zhejiang University of Medicine, Hangzhou, China
| | - Ke Wang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wei Wang
- Department of Pulmonary and Critical Care Medicine, the First Hospital of China Medical University, Shenyang, China
| | - Xiaoping Wang
- Department of Respiratory Disease, China-Japan Friendship Hospital, Beijing, China
| | - Yuehong Wang
- Department of Respiratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijie Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zirui Wang
- Department of Respiratory and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Lin Wu
- Thoracic Medicine Department II, Hunan Cancer Hospital, Changsha, China
| | - Di Wu
- Department of Respiratory Medicine, Shenzhen People’s Hospital, Shenzhen, China
| | - Baosong Xie
- Department of Respiratory Medicine, Fujian Provincial Hospital, Fuzhou, China
| | - Min Xie
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohong Xie
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Zhanhong Xie
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Shufeng Xu
- Department of Respiratory and Critical Care Medicine, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Xiaoman Xu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xia Yang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yan Yin
- Department of Pulmonary and Critical Care Medicine, the First Hospital of China Medical University, Shenyang, China
| | - Zongyang Yu
- Department of Pulmonary and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, China
| | - Jian Zhang
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jianqing Zhang
- Second Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jing Zhang
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingying Zhang
- Department of Medical Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Diansheng Zhong
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiangdong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yanbin Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bo Zhu
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chenxi Zou
- Department of Respiratory and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Nanshan Zhong
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Jianxing He
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Chunxue Bai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chengping Hu
- Department of Pulmonary Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing, China
| | - Jianying Zhou
- Department of Respiratory Diseases, The First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou, China
| | - Baohui Han
- Department of Pulmonology, Shanghai Chest Hospital, Shanghai, China
| | - Janos Varga
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Esther Barreiro
- Pulmonology Department-Lung Cancer and Muscle Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Department of Medicine and Life Sciences (MELIS), Pompeu Fabra University (UPF), CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII) Barcelona, Spain
| | - Hye Yun Park
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Francesco Petrella
- Division of Thoracic Surgery, IRCCS European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Yuichi Saito
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Taichiro Goto
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, Yamanashi, Japan
| | - Hitoshi Igai
- Department of General Thoracic Surgery, Japanese Red Cross Maebashi Hospital, Maebashi, Gunma, Japan
| | - Sara Bravaccini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Michele Zanoni
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Piergiorgio Solli
- Department of Cardio-Thoracic Surgery and Hearth & Lung Transplantation, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Alfonso Fiorelli
- Thoracic Surgery Unit, Universitàdella Campania Luigi Vanvitelli, Naples, Italy
| | - Takeo Nakada
- Division of Thoracic Surgery, Department of Surgery, the Jikei University School of Medicine, Tokyo, Japan
| | - Yoshinobu Ichiki
- Department of General Thoracic Surgery, Saitama Medical University International Medical Center, Saitama, Japan
| | - Rossana Berardi
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | | | - Nicolas Girard
- Institut du Thorax Curie Montsouris, Institut Curie, Paris, France
- Paris Saclay, UVSQ, Versailles, France
| | - Antonio Rossi
- Oncology Center of Excellence, Therapeutic Science & Strategy Unit, IQVIA, Milan, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Toyoaki Hida
- Lung Cancer Center, Central Japan International Medical Center, Minokamo, Japan
| | - Shiyue Li
- The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Liang’an Chen
- Department of Respiratory and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Rongchang Chen
- Shenzhen Institute of Respiratory Diseases, Shenzhen People’s Hospital, Shenzhen, China
- Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
21
|
Esparcia-Pinedo L, Romero-Laorden N, Alfranca A. Tertiary lymphoid structures and B lymphocytes: a promising therapeutic strategy to fight cancer. Front Immunol 2023; 14:1231315. [PMID: 37622111 PMCID: PMC10445545 DOI: 10.3389/fimmu.2023.1231315] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Tertiary lymphoid structures (TLSs) are clusters of lymphoid cells with an organization that resembles that of secondary lymphoid organs. Both structures share common developmental characteristics, although TLSs usually appear in chronically inflamed non-lymphoid tissues, such as tumors. TLSs contain diverse types of immune cells, with varying degrees of spatial organization that represent different stages of maturation. These structures support both humoral and cellular immune responses, thus the correlation between the existence of TLS and clinical outcomes in cancer patients has been extensively studied. The finding that TLSs are associated with better prognosis in some types of cancer has led to the design of therapeutic strategies based on promoting the formation of these structures. Agents such as chemokines, cytokines, antibodies and cancer vaccines have been used in combination with traditional antitumor treatments to enhance TLS generation, with good results. The induction of TLS formation therefore represents a novel and promising avenue for the treatment of a number of tumor types.
Collapse
Affiliation(s)
- Laura Esparcia-Pinedo
- Immunology Department, Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Department of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Nuria Romero-Laorden
- Medical Oncology Department, Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Cátedra Universidad Autónoma de Madrid (UAM)-Fundación Instituto Roche de Medicina Personalizada de Precisión, Madrid, Spain
| | - Arantzazu Alfranca
- Immunology Department, Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Department of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Cátedra Universidad Autónoma de Madrid (UAM)-Fundación Instituto Roche de Medicina Personalizada de Precisión, Madrid, Spain
- Centro de Investigación Biomédica en Red Cardiovascular, CIBERCV, Madrid, Spain
| |
Collapse
|
22
|
Koti M, Bivalacqua T, Black PC, Cathomen T, Galsky MD, Gulley JL, Ingersoll MA, Kamat AM, Kassouf W, Siemens DR, Gao J. Adaptive Immunity in Genitourinary Cancers. Eur Urol Oncol 2023; 6:263-272. [PMID: 37069029 DOI: 10.1016/j.euo.2023.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/21/2023] [Accepted: 03/09/2023] [Indexed: 04/19/2023]
Abstract
CONTEXT While urothelial and renal cell cancers have exhibited modest responses to novel immune checkpoint inhibitors targeting the programmed death ligand 1 and its receptor, response rates in patients with prostate cancer have remained poor. The factors underlying suboptimal outcomes observed in patients treated with novel immunotherapies are still to be resolved. OBJECTIVE To review the literature and describe the key adaptive immune physiological events associated with cancer progression and therapeutic response in genitourinary (GU) cancers. EVIDENCE ACQUISITION We performed a nonsystematic, collaborative narrative review to highlight recent advancements leading to the current state of knowledge on the critical mediators of antitumor adaptive immunity to GU cancers. Further, we discuss the findings on the pre- and post-treatment immunological events that either are unique to each of the three cancer types or exhibit overlapping clinical associations. EVIDENCE SYNTHESIS Aging-associated immune function decline is a major factor underlying poor outcomes observed in patients treated with both conventional and novel immunotherapies. Other cancer immunobiological aspects associated with suboptimal responses in GU cancers include the overall tumor mutational burden, mutations in specific tumor suppressor/DNA damage repair genes (KDM6A, PTEN, STAG2, TP53, ATM, and BRCA2), and abundance of multiple functional states of adaptive immune cells and their spatiotemporal localization within the tumor immune microenvironment. Understanding these mechanisms may potentially lead to the development of prognostic and predictive biomarkers such as immune cell infiltration profiles and tertiary lymphoid structures (TLSs) that associate with variable clinical outcomes depending on the nature of the novel immunotherapeutic approach. Implementation of newer immune-monitoring technologies and improved preclinical modeling systems will augment our understanding of the host and tumor intrinsic factors contributing to the variability of responses to immunotherapies. CONCLUSIONS Despite the tremendous progress made in the understanding of dynamic and static adaptive immune elements within the tumor immune landscape, several knowledge gaps remain. A comprehensive knowledge thus gained will lead to precision immunotherapy, improved drug sequencing, and a therapeutic response. PATIENT SUMMARY We performed a collaborative review by a diverse group of experts in the field to examine our understanding of the events and crosstalk between cancer cells and the patient's immune system that are associated with responses to novel immunotherapies. An evolving understanding of tumor-intrinsic and host-related immune alterations, both before and after therapy, will aid in the discovery of promising markers of responses to immunotherapy as well as the development of unique therapeutic approaches for the management of genitourinary cancers.
Collapse
Affiliation(s)
- Madhuri Koti
- Department of Biomedical and Molecular Sciences, Cancer Research Institute, Queen's University, Kingston, ON, Canada.
| | - Trinity Bivalacqua
- Department of Urology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter C Black
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Faculty of Medicine & Medical Center - University of Freiburg, Freiburg, Germany
| | - Matthew D Galsky
- Division of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James L Gulley
- Center for Immuno-Oncology, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Molly A Ingersoll
- Université Paris Cité, Institut Cochin, INSERM U1016, CNRS UMR 8104, Paris, 75014, France; Mucosal Inflammation and Immunity, Department of Immunology, Institut Pasteur, 75015 Paris, France
| | - Ashish M Kamat
- Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wassim Kassouf
- Division of Urology, McGill University Health Center, Montreal, QC, Canada
| | - D Robert Siemens
- Department of Urology, Queen's University School of Medicine, Kingston, ON, Canada
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
23
|
Shang T, Jiang T, Lu T, Wang H, Cui X, Pan Y, Xu M, Pei M, Ding Z, Feng X, Lin Y, Li X, Tan Y, Feng F, Dong H, Wang H, Dong L. Tertiary lymphoid structures predict the prognosis and immunotherapy response of cholangiocarcinoma. Front Immunol 2023; 14:1166497. [PMID: 37234171 PMCID: PMC10206168 DOI: 10.3389/fimmu.2023.1166497] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Cholangiocarcinoma (CCA) is a malignant tumor of the biliary epithelium with a poor prognosis. The lack of biomarkers to predict therapeutic response and prognosis is one of the major challenges for CCA treatment. Tertiary lymphoid structures (TLS) provide a local and pivotal microenvironment for tumor immune responses. The prognostic value and clinical relevance of TLS in CCA remain unclear. We aimed to explore the characteristics and clinical significance of TLS in CCA. Methods We investigated the prognostic value and clinical relevance of TLS in CCA using a surgery cohort containing 471 CCA patients (cohort 1) and an immunotherapy cohort containing 100 CCA patients (cohort 2). Hematoxylin and eosin (H&E) and immunohistochemical (IHC) staining were used to evaluate the maturity of TLS. Multiplex IHC (mIHC) was employed to characterize the composition of TLS. Results Different maturity of TLS were observed in CCA tissue sections. Strong staining of the four-gene signature including PAX5, TCL1A, TNFRSF13C, and CD79A were found in TLS regions. A high density of intra-tumoral TLS (T-score high) were significantly correlated with longer overall survival (OS) both in CCA cohort 1 (p = 0.002) and cohort 2 (p = 0.01), whereas a high density of peri-tumoral TLS (P-score high) were associated with shorter OS in these two cohorts (p = 0.003 and p = 0.03, respectively). Conclusion The established four-gene signature efficiently identified the TLS in CCA tissues. The abundance and spatial distribution of TLS were significantly correlated with the prognosis and immune checkpoint inhibitors (ICIs) immunotherapy response of CCA patients. The presence of intra-tumoral TLS are positive prognostic factors for CCA, which provide a theoretical basis for the future diagnosis and treatment of CCA.
Collapse
Affiliation(s)
- Taiyu Shang
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
- School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Tianyi Jiang
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Tao Lu
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Hui Wang
- Department of Hepatobiliary Diseases, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Xiaowen Cui
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Yufei Pan
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Mengyou Xu
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Mengmiao Pei
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Zhiwen Ding
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Xiaofan Feng
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Yunkai Lin
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Xin Li
- Department of Hepatobiliary Diseases, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Yexiong Tan
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Feiling Feng
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Hui Dong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Hongyang Wang
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
- School of Life Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Liwei Dong
- National Center for Liver Cancer, Naval Medical University, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| |
Collapse
|
24
|
Marquina Escalante F, Lévano Díaz C, Fuster Guillén D. [New therapeutic advances in patients with lung cancer immunosuppressed with chronic lung diseases in the period 2014-2022 from the review of the literature.]. Rev Esp Salud Publica 2023; 97:e202302015. [PMID: 37057359 PMCID: PMC10541256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/26/2023] [Indexed: 04/15/2023] Open
Abstract
Lung cancer is a malignant neoplasm with a high prevalence and mortality, more so in patients with respiratory comorbidities, whose cells have a massive proliferation capacity in the lung tissue, managing to invade other organs, which deteriorates the patient's physical and emotional state, decreasing their quality of life and defense system; therefore, treatment today is not sufficient for patient survival and there has been evidence of a certain evolution in the treatment of the disease or early detection to prevent it. This article aimed to analyze the new therapeutic advances in patients with lung cancer associated with chronic lung diseases in the period 2014-2022 based on a review of the literature. Several parameters were used to limit the search, extrapolating the articles of interest, validating fifty three articles, six doctoral theses and two books, which were in Spanish and English.The various search strategies used were keywords, subject and author follow-up. The sections developed in this review are the concept of Lung Cancer (LC), clinical manifestations, risk factors, relationship between LC and chronic lung diseases, diagnosis, treatment, prevention and new therapeutic advances. All the filtered information of the selected articles shows us the importance that the use of various biomarkers is taking for its early detection; however, the transfer of antitumor T cells in patients with underlying lung disease had an efficiency of 48.
Collapse
Affiliation(s)
- Fiorella Marquina Escalante
- Escuela de Medicina Humana, Universidad Privada San Juan BautistaUniversidad Privada San Juan BautistaChorrillosPerú
| | - César Lévano Díaz
- Escuela de Medicina Humana, Universidad Privada San Juan BautistaUniversidad Privada San Juan BautistaChorrillosPerú
| | - Doris Fuster Guillén
- Escuela de Medicina Humana, Universidad Privada San Juan BautistaUniversidad Privada San Juan BautistaChorrillosPerú
| |
Collapse
|
25
|
Wang Z, Shi X, Zhao Y, Zhou J, Zhang S, Wang J, Yu W, Zhang X, Ren X, Zhao H. DC101, an anti-VEGFR2 agent, promotes high-endothelial venule formation and immune infiltration versus SAR131675 and fruquintinib. Biochem Biophys Res Commun 2023; 661:10-20. [PMID: 37084488 DOI: 10.1016/j.bbrc.2023.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 04/08/2023] [Indexed: 04/23/2023]
Abstract
There is an increasing interest in combining immune checkpoint inhibitors (ICIs) with anti-angiogenic drugs to enhance their anti-tumor effects. In this study, three anti-angiogenic agents, DC101 (acting on VEGFR2), SAR131675 (acting on VEGFR3), and fruquintinib (a small-molecule inhibitor acting on multiple targets) were administered to B16F1-OVA-loaded C57BL/6 mice. Immune cells infiltration in the tumor tissues, vascular normalization, and high-endothelial venule (HEV) formation were assessed to provide evidence for drug combination. Both DC101 and fruquintinib significantly slowed the melanoma growth and increased the proportion of CD3+ and CD8+ T cells infiltration compared with SAR131675, of note, the effect of DC101 was more pronounced. Moreover, DC101 and fruquintinib increased the interferon-γ and perforin levels, meanwhile, DC101 increased the granzyme B levels, whereas fruquintinib and SAR131675 did not. Only the fruquintinib-treated group showed decreased regulatory T cells infiltration. We found upregulation of PD-L1 expression in tumor cells and CD45+ immune cells in DC101-treated group as well as upregulation of PD-1 expression on CD3+ T cells. However, fruquintinib only increased PD-L1 expression in tumors. Both DC101 and fruquintinib reduced the proportion of CD31+ vessels, while DC101 increased the ratio of α-SMA +/CD31+ cells and reduced the expression of HIF-1α more than fruquintinib. Moreover, DC101 enhanced the infiltration of dendritic cells and B cells, and local HEV formation. In conclusion, our data indicate that DC101 may be a better choice for the combined clinical application of ICIs and anti-angiogenic agents.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China; National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China
| | - Xiuhuan Shi
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China; National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China
| | - Yu Zhao
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China; National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China
| | - Jian Zhou
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China; National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China
| | - Siyuan Zhang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China; National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China
| | - Jiahui Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China; National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China
| | - Wenwen Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China; National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China
| | - Xiying Zhang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China; National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China; National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China; Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| | - Hua Zhao
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China; National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, China.
| |
Collapse
|
26
|
He M, He Q, Cai X, Liu J, Deng H, Li F, Zhong R, Lu Y, Peng H, Wu X, Chen Z, Lao S, Li C, Li J, He J, Liang W. Intratumoral tertiary lymphoid structure (TLS) maturation is influenced by draining lymph nodes of lung cancer. J Immunother Cancer 2023; 11:jitc-2022-005539. [PMID: 37072348 PMCID: PMC10124324 DOI: 10.1136/jitc-2022-005539] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Tertiary lymphoid structure (TLS) is an organized infiltration of immune cells, showing features of germinal center (GC) commonly seen in secondary lymphoid organs. However, its relationship with tumor-draining lymph nodes (TDLNs) has not been studied and we hypothesized that TDLN may influence maturation of intratumoral TLS in non-small cell lung cancer (NSCLC). METHODS Tissue slides of 616 patients that had undergone surgeries were examined. Cox proportional hazard regression model was used to assess risk factors of patients' survival, and logistic regression model was used for their relationship with TLS. Single-cell RNA-sequencing (scRNA-seq) was employed to explore transcriptomic features of TDLNs. Immunohistochemistry, multiplex immunofluorescence and flow cytometry were performed to analyze cellular composition. Cellular components of NSCLC samples from The Cancer Genome Atlas database were inferred with Microenvironment Cell Populations-counter (MCP-counter) method. Murine NSCLC models were used to dissect underlying mechanisms for relationship between TDLN and TLS maturation. RESULTS While GC+ TLS was associated with better prognosis, GC- TLS was not. TDLN metastasis reduced the prognostic relevance of TLS, and was associated with less GC formation. Primary tumor sites showed reduced B cell infiltration in TDLN-positive patients, and scRNA-seq revealed diminished memory B cell formation in tumor-invaded TDLNs, together with an emphasis on weakened interferon (IFN)-γ response. Murine NSCLC models revealed that IFN-γ signaling is involved in memory B cell differentiation in TDLNs and GC formation in primary tumors. CONCLUSIONS Our research emphasizes the influence of TDLN on intratumoral TLS maturation and suggests a role of memory B cells and IFN-γ signaling in this communication.
Collapse
Affiliation(s)
- Miao He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Qihua He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Xiuyu Cai
- Department of General Internal Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jun Liu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Hongshen Deng
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Feng Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Ran Zhong
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Yi Lu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Haoxin Peng
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Xiangrong Wu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Zisheng Chen
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Shen Lao
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Caichen Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Jianfu Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
- Department of surgery, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
- Department of Medical Oncology, The First People Hospital of Zhaoqing, Zhaoqing, Guangdong, China
| |
Collapse
|
27
|
Yang Y, Liu H, Chen Y, Xiao N, Zheng Z, Liu H, Wan J. Liquid biopsy on the horizon in immunotherapy of non-small cell lung cancer: current status, challenges, and perspectives. Cell Death Dis 2023; 14:230. [PMID: 37002211 PMCID: PMC10066332 DOI: 10.1038/s41419-023-05757-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 04/03/2023]
Abstract
Non-small cell lung cancer (NSCLC) is one of the most threatening malignancies to human health and life. In most cases, patients with NSCLC are already at an advanced stage when they are diagnosed. In recent years, lung cancer has made great progress in precision therapy, but the efficacy of immunotherapy is unstable, and its response rate varies from patient to patient. Several biomarkers have been proposed to predict the outcomes of immunotherapy, such as programmed cell death-ligand 1 (PD-L1) expression and tumor mutational burden (TMB). Nevertheless, the detection assays are invasive and demanding on tumor tissue. To effectively predict the outcomes of immunotherapy, novel biomarkers are needed to improve the performance of conventional biomarkers. Liquid biopsy is to capture and detect circulating tumor cells (CTCs), circulating tumor DNA (ctDNA) and exosomes in body fluids, such as blood, saliva, urine, pleural fluid and cerebrospinal fluid as samples from patients, so as to make analysis and diagnosis of cancer and other diseases. The application of liquid biopsy provides a new possible solution, as it has several advantages such as non-invasive, real-time dynamic monitoring, and overcoming tumor heterogeneity. Liquid biopsy has shown predictive value in immunotherapy, significantly improving the precision treatment of lung cancer patients. Herein, we review the application of liquid biopsy in predicting the outcomes of immunotherapy in NSCLC patients, and discuss the challenges and future directions in this field.
Collapse
Affiliation(s)
- Ying Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Youming Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Nan Xiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhaoyang Zheng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Hongchun Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
28
|
Peng H, Wu X, Liu S, He M, Xie C, Zhong R, Liu J, Tang C, Li C, Xiong S, Zheng H, He J, Lu X, Liang W. Multiplex immunofluorescence and single-cell transcriptomic profiling reveal the spatial cell interaction networks in the non-small cell lung cancer microenvironment. Clin Transl Med 2023; 13:e1155. [PMID: 36588094 PMCID: PMC9806015 DOI: 10.1002/ctm2.1155] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Conventional immunohistochemistry technologies were limited by the inability to simultaneously detect multiple markers and the lack of identifying spatial relationships among cells, hindering understanding of the biological processes in cancer immunology. METHODS Tissue slices of primary tumours from 553 IA∼IIIB non-small cell lung cancer (NSCLC) cases were stained by multiplex immunofluorescence (mIF) assay for 10 markers, including CD4, CD38, CD20, FOXP3, CD66b, CD8, CD68, PD-L1, CD133 and CD163, evaluating the amounts of 26 phenotypes of cells in tumour nest and tumour stroma. StarDist depth learning model was utilised to determine the spatial location of cells based on mIF graphs. Single-cell RNA sequencing (scRNA-seq) on four primary NSCLC cases was conducted to investigate the putative cell interaction networks. RESULTS Spatial proximity among CD20+ B cells, CD4+ T cells and CD38+ T cells (r2 = 0.41) was observed, whereas the distribution of regulatory T cells was associated with decreased infiltration levels of CD20+ B cells and CD38+ T cells (r2 = -0.45). Univariate Cox analyses identified closer proximity between CD8+ T cells predicted longer disease-free survival (DFS). In contrast, closer proximity between CD133+ cancer stem cells (CSCs), longer distances between CD4+ T cells and CD20+ B cells, CD4+ T cells and neutrophils, and CD20+ B cells and neutrophils were correlated with dismal DFS. Data from scRNA-seq further showed that spatially adjacent N1-like neutrophils could boost the proliferation and activation of T and B lymphocytes, whereas spatially neighbouring M2-like macrophages showed negative effects. An immune-related risk score (IRRS) system aggregating robust quantitative and spatial prognosticators showed that high-IRRS patients had significantly worse DFS than low-IRRS ones (HR 2.72, 95% CI 1.87-3.94, p < .001). CONCLUSIONS We developed a framework to analyse the cell interaction networks in tumour microenvironment, revealing the spatial architecture and intricate interplays between immune and tumour cells.
Collapse
Affiliation(s)
- Haoxin Peng
- Department of Thoracic Oncology and SurgeryChina State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Clinical MedicineNanshan SchoolGuangzhou Medical UniversityGuangzhouChina
| | - Xiangrong Wu
- Department of Thoracic Oncology and SurgeryChina State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Clinical MedicineNanshan SchoolGuangzhou Medical UniversityGuangzhouChina
| | - Shaopeng Liu
- Department of Computer ScienceGuangdong Polytechnic Normal UniversityGuangzhouChina
- Department of Artificial Intelligence ResearchPazhou LabGuangzhouChina
| | - Miao He
- Department of Thoracic Oncology and SurgeryChina State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Chao Xie
- Department of Computer ScienceGuangdong Polytechnic Normal UniversityGuangzhouChina
| | - Ran Zhong
- Department of Thoracic Oncology and SurgeryChina State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Jun Liu
- Department of Thoracic Oncology and SurgeryChina State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Chenshuo Tang
- Department of Computer ScienceGuangdong Polytechnic Normal UniversityGuangzhouChina
| | - Caichen Li
- Department of Thoracic Oncology and SurgeryChina State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Shan Xiong
- Department of Thoracic Oncology and SurgeryChina State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Hongbo Zheng
- Medical DepartmentGenecast Biotechnology Co., LtdBeijingChina
| | - Jianxing He
- Department of Thoracic Oncology and SurgeryChina State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Xu Lu
- Department of Computer ScienceGuangdong Polytechnic Normal UniversityGuangzhouChina
- Department of Artificial Intelligence ResearchPazhou LabGuangzhouChina
| | - Wenhua Liang
- Department of Thoracic Oncology and SurgeryChina State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Medical OncologyThe First People's Hospital of ZhaoqingZhaoqingChina
| |
Collapse
|
29
|
Tertiary Lymphoid Structures: A Potential Biomarker for Anti-Cancer Therapy. Cancers (Basel) 2022; 14:cancers14235968. [PMID: 36497450 PMCID: PMC9739898 DOI: 10.3390/cancers14235968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
A tertiary lymphoid structure (TLS) is a special component in the immune microenvironment that is mainly composed of tumor-infiltrating lymphocytes (TILs), including T cells, B cells, DC cells, and high endothelial venules (HEVs). For cancer patients, evaluation of the immune microenvironment has a predictive effect on tumor biological behavior, treatment methods, and prognosis. As a result, TLSs have begun to attract the attention of researchers as a new potential biomarker. However, the composition and mechanisms of TLSs are still unclear, and clinical detection methods are still being explored. Although some meaningful results have been obtained in clinical trials, there is still a long way to go before such methods can be applied in clinical practice. However, we believe that with the continuous progress of basic research and clinical trials, TLS detection and related treatment can benefit more and more patients. In this review, we generalize the definition and composition of TLSs, summarize clinical trials involving TLSs according to treatment methods, and describe possible methods of inducing TLS formation.
Collapse
|
30
|
The impact of tertiary lymphoid structures on clinicopathological, genetic and gene expression characteristics in lung adenocarcinoma. Lung Cancer 2022; 174:125-132. [PMID: 36379125 DOI: 10.1016/j.lungcan.2022.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/14/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Tertiary lymphoid structures (TLS) are observed in several cancers and are associated with favorable prognosis. This study aimed to examine the clinicopathological, genetic, and gene expression profiles of lung adenocarcinoma patients with TLS. METHODS A total of 112 patients with pathological stage IB lung adenocarcinoma who underwent complete resection between 2011 and 2015 were enrolled in this study. We investigated whether TLS correlated with prognosis and programmed death-ligand 1 (PD-L1) expression. Furthermore, the correlation of TLS with tumor mutation burden (TMB) and genetic mutations was evaluated in patients for whom whole-exon sequencing data were available. In addition, using the Cancer Genome Atlas Lung Adenocarcinoma (TCGA-LUAD) dataset, gene expression analysis according to the TLS status was performed. RESULTS Among the 112 patients, 49 were TLS-positive (TLS+). TLS+ correlated with longer recurrence-free survival (RFS) than TLS-negative cases (TLS-) (hazard ratio [HR], 0.47; 95 % confidence interval [CI]: 0.23-0.88, p = 0.02). In the multivariate analysis, TLS was a better independent prognostic factor for RFS (HR 0.37, 95 %CI 0.18-0.72, p < 0.01). PD-L1 expression was not significantly different between TLS+ and TLS- patients (p = 0.54). TMB in TLS+ was similar to that in TLS- patients (p = 0.39); however, it tended to be lower than that in TLS- especially among smokers (p = 0.07). In gene expression analysis, RNA expression of chemokines related to lymph node formation, such as CXCL13, CCL19 and CCL21, was significantly higher, and biological processes such as positive regulation of humoral immune response and regulation of antigen receptor-mediated signaling pathway were enhanced in TLS+. CONCLUSIONS TLS was a favorable prognostic factor and was not associated with PD-L1 expression in patients with lung adenocarcainoma. Moreover, gene expression analysis indicated that TLS is a site for the generation and regulation of antitumor immune responses.
Collapse
|
31
|
Wang F, Yang M, Luo W, Zhou Q. Characteristics of tumor microenvironment and novel immunotherapeutic strategies for non-small cell lung cancer. JOURNAL OF THE NATIONAL CANCER CENTER 2022; 2:243-262. [PMID: 39036549 PMCID: PMC11256730 DOI: 10.1016/j.jncc.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/08/2022] Open
Abstract
Immune checkpoint inhibitor-based immunotherapy has revolutionized the treatment approach of non-small cell lung cancer (NSCLC). Monoclonal antibodies against programmed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) are widely used in clinical practice, but other antibodies that can circumvent innate and acquired resistance are bound to undergo preclinical and clinical studies. However, tumor cells can develop and facilitate the tolerogenic nature of the tumor microenvironment (TME), resulting in tumor progression. Therefore, the immune escape mechanisms exploited by growing lung cancer involve a fine interplay between all actors in the TME. A better understanding of the molecular biology of lung cancer and the cellular/molecular mechanisms involved in the crosstalk between lung cancer cells and immune cells in the TME could identify novel therapeutic weapons in the old war against lung cancer. This article discusses the role of TME in the progression of lung cancer and pinpoints possible advances and challenges of immunotherapy for NSCLC.
Collapse
Affiliation(s)
- Fen Wang
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Mingyi Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Weichi Luo
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
32
|
Effect of Tertiary Lymphoid Structures on Prognosis of Patients with Hepatocellular Carcinoma and Preliminary Exploration of Its Formation Mechanism. Cancers (Basel) 2022; 14:cancers14205157. [PMID: 36291944 PMCID: PMC9601110 DOI: 10.3390/cancers14205157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 11/18/2022] Open
Abstract
Simple Summary At present, research on tertiary lymphoid structures (TLSs) in hepatocellular carcinoma (HCC) has been limited to the prognostic impact. Our manuscript first validates previous studies using two databases and then initially explores the key molecules and mechanisms of TLS formation and immunotherapy implications for HCC patients by using the TCGA database. For example, LCK, a key molecule in the formation of TLSs, may affect the formation of TLSs by regulating the cytokine signalling pathway, chemokine signalling pathway, T-cell activation and P53 signalling pathway. Second, the expression level of LCK is another factor affecting the sensitivity of HCC patients to immune checkpoint inhibitors. In conclusion, our study provides a potential mechanism for further exploration of TLSs. Abstract Background: Tertiary lymphoid structures (TLSs) are formed by the aggregation of tumour-infiltrating lymphocytes (TILs), which is driven by chemokines or cytokines in the tumour microenvironment. Studies have shown that TLSs are associated with good prognosis in patients with various solid tumours and can improve patient responses to immunotherapy. However, the role of TLSs in hepatocellular carcinoma (HCC) remains controversial, and the underlying molecular mechanism is unclear. Methods: According to haematoxylin-eosin (HE) staining results, HCC patients in Xijing Hospital data and TCGA data were divided into TLS+ and TLS- groups, and Kaplan–Meier (KM) analysis was performed to assess overall survival (OS) and recurrence-free survival (RFS). Immunofluorescence (IF) and immunohistochemistry (IHC) were used to identify TILs in the TLS+ group. Lymphocyte-specific protein tyrosine kinase (LCK), a molecule involved in TLS formation, was explored in LinkedOmics. TILs were divided into two groups by drawing receiver operating characteristic (ROC) curves to calculate cut-off values. Spearman correlation analysis was used to calculate the correlation between LCK and TILs, and the molecular pathways by which LCK regulates immunotherapy were clarified through enrichment analysis. The half-maximal inhibitory concentration (IC50) distribution of sorafenib was observed in groups that varied in LCK expression. Results: According to the HE results, 61 cases in the Xijing Hospital cohort and 195 cases in the TCGA cohort had TLSs, while 89 cases and 136 cases did not. The KM results showed that TLSs had no effect on the OS of HCC patients but significantly affected RFS. The IF/IHC results showed that higher TIL numbers in TLSs were correlated with better prognosis in HCC patients. Spearman correlation analysis showed that LCK expression was positively correlated with TIL numbers. Enrichment analysis showed that upregulation of LCK expression mainly regulated the cytokine signalling pathway, the chemokine signalling pathway and T-cell activation. The IC50 scores of sorafenib in HCC patients with high LCK expression were lower, and the sensitivity was higher. Conclusion: TLSs mainly affected the early RFS of HCC patients but had no effect on OS. The high expression of the TLS formation-related gene LCK can increase the sensitivity of HCC patients to ICIs.
Collapse
|
33
|
Ren F, Xie M, Gao J, Wu C, Xu Y, Zang X, Ma X, Deng H, Song J, Huang A, Pang L, Qian J, Yu Z, Zhuang G, Liu S, Pan L, Xue X. Tertiary lymphoid structures in lung adenocarcinoma: characteristics and related factors. Cancer Med 2022; 11:2969-2977. [PMID: 35801360 PMCID: PMC9359870 DOI: 10.1002/cam4.4796] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/15/2021] [Accepted: 12/18/2021] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Tertiary lymphoid structures (TLSs) are found in a variety of malignancies and affect the growth of tumors, but few studies have addressed their role in lung adenocarcinoma (LAC). We aimed to evaluate clinical features associated with TLSs in patients with LAC. METHODS AND MATERIALS A collection of resected pulmonary nodules in patients with LAC was retrospectively analyzed. TLSs were quantified by their number per square millimeter tumor area (density) and by the degree of lymphocyte aggregation (maturity) in each case. The correlation between TLS density and maturity and clinical features was calculated. RESULTS A total of 243 patients were selected, of whom 219 exhibited TLSs. The occurrence of TLSs was correlated with computed tomography (CT) features as follows: pure ground-glass nodules (pGGNs) (n = 43) was associated with a lower occurrence rate than part-solid nodules (PSNs) (n = 112) and solid nodules (SNs) were (n = 88) (p = 0.037). TLS density was correlated with age and CT features. Poisson regression showed higher TLS density in PSNs and SNs than in pGGNs (incidence rate ratio [IRR]: 3.137; 95% confidence interval [CI]: 1.35-7.27; p = 0.008 and IRR: 2.44; 95% CI: 1.02-5.85; p = 0.046, respectively). In addition, TLS density was higher in patients aged under 60 years than in those aged over 60 years (IRR: 0.605; 95% CI: 0.4-0.92; p = 0.018). The maturity of TLSs was higher in patients with higher tumor stages (p = 0.026). CONCLUSIONS We demonstrated distinct profiles of TLSs in early LAC and their correlations with CT features, age, and tumor stages, which could help understand tumor progression and management.
Collapse
Affiliation(s)
- Fangping Ren
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, Beijing, P.R. China
| | - Mei Xie
- Department of Respiratory and Critical Care, the Chinese PLA General Hospital, Beijing, P.R. China
| | - Jie Gao
- Department of Pathology, the Chinese PLA General Hospital, Beijing, P.R. China
| | - Chongchong Wu
- Department of Radiology, the Chinese PLA General Hospital, Beijing, P.R. China
| | - Yang Xu
- Department of Respiratory and Critical Care, the Chinese PLA General Hospital, Beijing, P.R. China
| | - Xuelei Zang
- Center of Clinical Laboratory Medicine, the first Medical Centre, Chinese PLA General Hospital, Beijing, P.R. China
| | - Xidong Ma
- Department of Respiratory and Critical Care, the Chinese PLA General Hospital, Beijing, P.R. China
| | - Hui Deng
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, Beijing, P.R. China
| | - Jialin Song
- Department of Respiratory Medicine, Weifang Medical university, Weifang, People's Republic of China
| | - Aiben Huang
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, Beijing, P.R. China
| | - Li Pang
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, Beijing, P.R. China
| | - Jin Qian
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, Beijing, P.R. China
| | - Zhaofeng Yu
- School of Medicine, Peking University, Beijing, P.R. China
| | - Guanglei Zhuang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Sanhong Liu
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, P.R. China
| | - Lei Pan
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, Beijing, P.R. China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
34
|
Histopathological Tissue Segmentation of Lung Cancer with Bilinear CNN and Soft Attention. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7966553. [PMID: 35845926 PMCID: PMC9283032 DOI: 10.1155/2022/7966553] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/15/2022] [Accepted: 06/10/2022] [Indexed: 11/18/2022]
Abstract
Automatic tissue segmentation in whole-slide images (WSIs) is a critical task in hematoxylin and eosin- (H&E-) stained histopathological images for accurate diagnosis and risk stratification of lung cancer. Patch classification and stitching the classification results can fast conduct tissue segmentation of WSIs. However, due to the tumour heterogeneity, large intraclass variability and small interclass variability make the classification task challenging. In this paper, we propose a novel bilinear convolutional neural network- (Bilinear-CNN-) based model with a bilinear convolutional module and a soft attention module to tackle this problem. This method investigates the intraclass semantic correspondence and focuses on the more distinguishable features that make feature output variations relatively large between interclass. The performance of the Bilinear-CNN-based model is compared with other state-of-the-art methods on the histopathological classification dataset, which consists of 107.7 k patches of lung cancer. We further evaluate our proposed algorithm on an additional dataset from colorectal cancer. Extensive experiments show that the performance of our proposed method is superior to that of previous state-of-the-art ones and the interpretability of our proposed method is demonstrated by Grad-CAM.
Collapse
|
35
|
Xia Y, Zha J, Curull V, Sánchez-Font A, Guitart M, Rodríguez-Fuster A, Aguiló R, Barreiro E. Gene expression profile of epithelial-mesenchymal transition in tumors of patients with nsclc: the influence of COPD. ERJ Open Res 2022; 8:00105-2022. [PMID: 35854873 PMCID: PMC9289374 DOI: 10.1183/23120541.00105-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/10/2022] [Indexed: 11/28/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT) is involved in the pathophysiology of lung cancer (LC) and COPD, and the latter is an important risk factor for LC. We hypothesised that the EMT gene expression profile and signalling cascade may differ in LC patients with COPD from those with no respiratory diseases. In lung tumour specimens obtained through video-assisted thoracoscopic surgery from LC (n=20, control group) and LC-COPD patients (n=30), gene expression (quantitative real-time PCR amplification) of EMT markers SMAD3, SMAD4, ZEB2, TWIST1, SNAI1, ICAM1, VIM, CDH2, MMP1 and MMP9 was detected. In lung tumours of LC-COPD compared to LC patients, gene expression of SMAD3, SMAD4, ZEB2 and CDH2 significantly declined, while no significant differences were detected for the other analysed markers. A significant correlation was found between pack-years (smoking burden) and SMAD3 gene expression among LC-COPD patients. LC-COPD patients exhibited mild-to-moderate airway obstruction and a significant reduction in diffusion capacity compared to LC patients. In lung tumour samples of patients with COPD, several markers of EMT expression, namely SMAD3, SMAD4, ZEB2 and CDH2, were differentially expressed suggesting that these markers are likely to play a role in the regulation of EMT in patients with this respiratory disease. Cigarette smoke did not seem to influence the expression of EMT markers in this study. These results have potential clinical implications in the management of patients with LC, particularly in those with underlying respiratory diseases. The downregulation of the epithelial–mesenchymal transition repressor SMAD pathway may favour a pro-tumoural micro-environment in patients with chronic airway diseases, namely COPD, which could be targeted therapeuticallyhttps://bit.ly/39oXnoG
Collapse
|
36
|
Li H, Li G, Xu P, Li Z. B cells and tumor immune escape. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:358-363. [PMID: 35545329 PMCID: PMC10930053 DOI: 10.11817/j.issn.1672-7347.2022.210275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Indexed: 06/15/2023]
Abstract
B lymphocyte is an important component of the human immune system and it has a role in the process of the body's specific immunity. In recent years, the research on B cells and tumor immune escape has rapidly progressed. Studies have shown that different types of B cells play different roles in tumor microenvironment through a variety of mechanisms. B cells in the tertiary lymphatic structure promote anti-tumor immunity, while regulatory B cells promote tumor immune escape. Antibody drugs targeting B cells are a promising direction for tumor immunotherapy.
Collapse
Affiliation(s)
- Huiting Li
- Cancer Research Institute, Central South University, Changsha 410078.
| | - Guiyuan Li
- Cancer Research Institute, Central South University, Changsha 410078
| | - Ping Xu
- Departments of Respiratory and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen Guangdong 518036, China
| | - Zheng Li
- Cancer Research Institute, Central South University, Changsha 410078.
| |
Collapse
|
37
|
Genova C, Dellepiane C, Carrega P, Sommariva S, Ferlazzo G, Pronzato P, Gangemi R, Filaci G, Coco S, Croce M. Therapeutic Implications of Tumor Microenvironment in Lung Cancer: Focus on Immune Checkpoint Blockade. Front Immunol 2022; 12:799455. [PMID: 35069581 PMCID: PMC8777268 DOI: 10.3389/fimmu.2021.799455] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
In the last decade, the treatment of non-small cell lung cancer (NSCLC) has been revolutionized by the introduction of immune checkpoint inhibitors (ICI) directed against programmed death protein 1 (PD-1) and its ligand (PD-L1), or cytotoxic T lymphocyte antigen 4 (CTLA-4). In spite of these improvements, some patients do not achieve any benefit from ICI, and inevitably develop resistance to therapy over time. Tumor microenvironment (TME) might influence response to immunotherapy due to its prominent role in the multiple interactions between neoplastic cells and the immune system. Studies investigating lung cancer from the perspective of TME pointed out a complex scenario where tumor angiogenesis, soluble factors, immune suppressive/regulatory elements and cells composing TME itself participate to tumor growth. In this review, we point out the current state of knowledge involving the relationship between tumor cells and the components of TME in NSCLC as well as their interactions with immunotherapy providing an update on novel predictors of benefit from currently employed ICI or new therapeutic targets of investigational agents. In first place, increasing evidence suggests that TME might represent a promising biomarker of sensitivity to ICI, based on the presence of immune-modulating cells, such as Treg, myeloid derived suppressor cells, and tumor associated macrophages, which are known to induce an immunosuppressive environment, poorly responsive to ICI. Consequently, multiple clinical studies have been designed to influence TME towards a pro-immunogenic state and subsequently improve the activity of ICI. Currently, the mostly employed approach relies on the association of "classic" ICI targeting PD-1/PD-L1 and novel agents directed on molecules, such as LAG-3 and TIM-3. To date, some trials have already shown promising results, while a multitude of prospective studies are ongoing, and their results might significantly influence the future approach to cancer immunotherapy.
Collapse
Affiliation(s)
- Carlo Genova
- UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università degli Studi di Genova, Genova, Italy
| | - Chiara Dellepiane
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Paolo Carrega
- Dipartimento di Patologia Umana, University of Messina, Messina, Italy
| | - Sara Sommariva
- SuPerconducting and Other INnovative Materials and Devices Institute, Consiglio Nazionale delle Ricerche (CNR-SPIN), Genova, Italy
- Life Science Computational Laboratory (LISCOMP), IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Guido Ferlazzo
- Dipartimento di Patologia Umana, University of Messina, Messina, Italy
| | - Paolo Pronzato
- UO Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Rosaria Gangemi
- UO Bioterapie, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Gilberto Filaci
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università degli Studi di Genova, Genova, Italy
- UO Bioterapie, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Simona Coco
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Michela Croce
- UO Bioterapie, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
38
|
Zhou X, Li W, Yang J, Qi X, Chen Y, Yang H, Chu L. Tertiary lymphoid structure stratifies glioma into three distinct tumor subtypes. Aging (Albany NY) 2021; 13:26063-26094. [PMID: 34954691 PMCID: PMC8751592 DOI: 10.18632/aging.203798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/11/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Tertiary lymphoid structure (TLS), also known as ectopic lymphoid organs, are found in cancer, chronic inflammation, and autoimmune diseases. However, the heterogeneity of TLS in gliomas is unclear. Therefore, it is necessary to identify TLS differences and define TLS subtypes. METHODS The TLS gene profile of 697 gliomas from The Cancer Genome Atlas (TCGA) was used for consensus clustering to identify robust clusters, and the reproducibility of the stratification method was assessed in Chinese Glioma Genome Atlas (CGGA) cohort1, CGGA_cohort2, and GSE16011. Analyses of clinical characteristics, immune infiltration, and potential biological functions were performed for each subtype. RESULTS Three resulting clusters (A, B, and C) were identified based on consensus clustering on the gene expression profile of TLS genes. There was a significant prognostic difference among the clusters, with a shorter survival for C than B and A. In comparison with the A and B subtypes, the C subtype was significantly enriched in primary immunodeficiency, intestinal immune network for lgG production, antigen processing and presentation, natural killer cell-mediated cytotoxicity, complement and coagulation cascades, cytokine-cytokine receptor interaction, leukocyte transendothelial migration, and some immune-related diseases. The levels of 23 immune cell types were higher in the C subtype than in the A and B subtypes. Finally, we developed and validated a riskscore based on TLS subtypes with better performance of prognosis prediction. CONCLUSIONS This study presents a new stratification method according to the TLS gene profile and highlights TLS heterogeneity in gliomas.
Collapse
Affiliation(s)
- Xingwang Zhou
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Wenyan Li
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Jie Yang
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Yimin Chen
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Hua Yang
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Liangzhao Chu
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| |
Collapse
|
39
|
Wang MY, Zhu WW, Zhang JY, Yu M, Zhai RD, Liu LK. Tertiary lymphoid structures in oral lichen planus and oral epithelial dysplasia with lichenoid features: A comparative study. Oral Dis 2021; 29:154-164. [PMID: 34897887 DOI: 10.1111/odi.14097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/13/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Tertiary lymphoid structures (TLSs) provide sites for antigen presentation and activation of lymphocytes, promoting their infiltration; thus, enhancing specific immune responses. The aim of this comparative cross-sectional study was to reveal the characteristics and influence of TLSs in oral lichen planus (OLP) and oral epithelial dysplasia (OED) with lichenoid features. METHODS Clinical information and samples of 51 OLP and 19 OED with lichenoid features were collected. Immunohistochemistry was performed, and the structures where CD20+ B cells and CD3+ T cells aggregated with peripheral lymph node addressin positive (PNAd+) vessels were defined as TLSs. The results and clinical information were analysed. RESULT TLS were found in 44 (86.3%) patients with OLP and 19 (100%) patients with OED. The TLS score was higher in OED group (p = 0.023), accompanied by an increased number of PNAd+ vessels. The TLS was significantly correlated with PNAd+ vessels (p = 0.027), CD20+ B (p < 0.001) and CD208+ dendritic cells (p = 0.001). Foxp3+ Treg cells but not CD8+ T cells infiltrated more severely in OED (p = 0.003) and increased when TLS score was high (p = 0.002). CONCLUSIONS This study revealed the widespread development of TLSs in the OLP and OED. The presence of TLSs showed a close relationship with dysplasia and may increase malignant potency by over-inducing Treg cells.
Collapse
Affiliation(s)
- Meng-Yao Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China.,Department of Basic Science of Stomatology, the affiliated Hospital of Stomatology, Nanjing Medical University, Jiangsu, China
| | - Wei-Wen Zhu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China.,Department of Basic Science of Stomatology, the affiliated Hospital of Stomatology, Nanjing Medical University, Jiangsu, China
| | - Jia-Yi Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China.,Department of Basic Science of Stomatology, the affiliated Hospital of Stomatology, Nanjing Medical University, Jiangsu, China
| | - Miao Yu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China.,Department of Periodontology, the affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Run-Dong Zhai
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China.,Department of Basic Science of Stomatology, the affiliated Hospital of Stomatology, Nanjing Medical University, Jiangsu, China
| | - Lai-Kui Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Jiangsu, China.,Department of Basic Science of Stomatology, the affiliated Hospital of Stomatology, Nanjing Medical University, Jiangsu, China
| |
Collapse
|
40
|
Aiba T, Hattori C, Sugisaka J, Shimizu H, Ono H, Domeki Y, Saito R, Kawana S, Kawashima Y, Terayama K, Toi Y, Nakamura A, Yamanda S, Kimura Y, Suzuki Y, Niida A, Sugawara S. Gene expression signatures as candidate biomarkers of response to PD-1 blockade in non-small cell lung cancers. PLoS One 2021; 16:e0260500. [PMID: 34843570 PMCID: PMC8629226 DOI: 10.1371/journal.pone.0260500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/10/2021] [Indexed: 11/18/2022] Open
Abstract
Although anti-PD-1/PD-L1 monotherapy has achieved clinical success in non-small cell lung cancer (NSCLC), definitive predictive biomarkers remain to be elucidated. In this study, we performed whole-transcriptome sequencing of pretreatment tumor tissue samples and pretreatment and on-treatment whole blood samples (WB) samples obtained from a clinically annotated cohort of NSCLC patients (n = 40) treated with nivolumab (anti-PD-1) monotherapy. Using a single-sample gene set enrichment scoring method, we found that the tumors of responders with lung adenocarcinoma (LUAD, n = 20) are inherently immunogenic to promote antitumor immunity, whereas those with lung squamous cell carcinoma (LUSC, n = 18) have a less immunosuppressive tumor microenvironment. These findings suggested that nivolumab may function as a molecular targeted agent in LUAD and as an immunomodulating agent in LUSC. In addition, our study explains why the reliability of PD-L1 expression on tumor cells as a predictive biomarker for the response to nivolumab monotherapy is quite different between LUAD and LUSC.
Collapse
Affiliation(s)
- Tomoiki Aiba
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
- * E-mail:
| | - Chieko Hattori
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Jun Sugisaka
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Hisashi Shimizu
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Hirotaka Ono
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yutaka Domeki
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Ryohei Saito
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Sachiko Kawana
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yosuke Kawashima
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Keisuke Terayama
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yukihiro Toi
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Atsushi Nakamura
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Shinsuke Yamanda
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yuichiro Kimura
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Atsushi Niida
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| |
Collapse
|
41
|
Gavrielatou N, Vathiotis I, Economopoulou P, Psyrri A. The Role of B Cells in Head and Neck Cancer. Cancers (Basel) 2021; 13:cancers13215383. [PMID: 34771546 PMCID: PMC8582491 DOI: 10.3390/cancers13215383] [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: 09/30/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 01/13/2023] Open
Abstract
Simple Summary Host immunity has established its role in deciding the course of cancer evolution. As cellular and molecular components in the tumor microenvironment peripherally appear to be at a constant interplay, favoring either tumor control or progression, it is vital to decrypt the immunity elements, which demonstrate the potential to be harnessed towards cancer elimination. Head and neck cancer has been characterized as densely immune infiltrated but at the same time a highly immunosuppressive malignancy due to a negative equilibrium between active and dysfunctional immune cell populations. B-cells constitute the cornerstone of humoral immunity; however, their role in cancer has been vastly overlooked in comparison to other immune subtypes and reports from multiple studies fail to show agreement on their prognostic impact. This review focuses on the role of B-cells on head and neck cancer with the aim to highlight their effect on anti-cancer immunity, as well as their possible impact on immunotherapy outcomes. Abstract Head and neck cancer comprises a heterogenous, highly immune infiltrated malignancy, defined by a predominantly immunosuppressive tumor microenvironment (TME). In recent years, PD-1/PD-L1 immune checkpoint inhibitors have become the standard of care treatment, either as monotherapy or in combination with chemotherapy agents, thus revolutionizing the therapeutic landscape of recurrent/metastatic disease. As a result, preclinical research is increasingly focusing on TME composition and pathophysiology, aiming to comprehensively characterize the specific elements and interactions affecting anti-tumor immunity, as well as to unveil novel predictive biomarkers of immunotherapy outcomes. While T lymphocytic populations have been vastly explored regarding their effect on cancer development, B-cells constitute a far less investigated, yet possibly equally important, aspect of cancer immunity. B-cell presence, either as single cells or as part of tertiary lymphoid structures within the TME, has been associated with several anti-tumor defense mechanisms, such as antigen presentation, antibody production and participation in antibody-dependent cellular cytotoxicity, and has demonstrated prognostic significance for multiple types of malignancies. However, immunoregulatory B-cell phenotypes have also been identified both peripherally and within malignant tissue, bearing inhibitory effects on numerous immune response processes. Consequently, B-cells and their subsets demonstrate the potential to become valuable cancer biomarkers and acquire a leading role in future therapeutic strategies.
Collapse
Affiliation(s)
- Niki Gavrielatou
- Department of Pathology, School of Medicine, Yale University, New Haven, CT 06510, USA; (N.G.); (I.V.)
| | - Ioannis Vathiotis
- Department of Pathology, School of Medicine, Yale University, New Haven, CT 06510, USA; (N.G.); (I.V.)
| | - Panagiota Economopoulou
- Section of Medical Oncology, Second Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Amanda Psyrri
- Section of Medical Oncology, Second Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
- Correspondence:
| |
Collapse
|
42
|
Qin L, Guitart M, Curull V, Sánchez-Font A, Duran X, Tang J, Admetlló M, Barreiro E. Systemic Profiles of microRNAs, Redox Balance, and Inflammation in Lung Cancer Patients: Influence of COPD. Biomedicines 2021; 9:biomedicines9101347. [PMID: 34680465 PMCID: PMC8533450 DOI: 10.3390/biomedicines9101347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 02/05/2023] Open
Abstract
Lung cancer (LC) risk increases in patients with chronic respiratory diseases (COPD). MicroRNAs and redox imbalance are involved in lung tumorigenesis in COPD patients. Whether systemic alterations of those events may also take place in LC patients remains unknown. Our objectives were to assess the plasma levels of microRNAs, redox balance, and cytokines in LC patients with/without COPD. MicroRNAs (RT-PCR) involved in LC, oxidized DNA, MDA-protein adducts, GSH, TEAC, VEGF, and TGF-beta (ELISA) were quantified in plasma samples from non-LC controls (n = 45), LC-only patients (n = 32), and LC-COPD patients (n = 91). In LC-COPD patients compared to controls and LC-only, MDA-protein adduct levels increased, while those of GSH decreased, and two patterns of plasma microRNA were detected. In both LC patient groups, miR-451 expression was downregulated, while those of microRNA-let7c were upregulated, and levels of TEAC and TGF-beta increased compared to the controls. Correlations were found between clinical and biological variables. A differential expression profile of microRNAs was detected in patients with LC. Moreover, in LC patients with COPD, plasma oxidative stress levels increased, whereas those of GSH declined. Systemic oxidative and antioxidant markers are differentially expressed in LC patients with respiratory diseases, thus implying its contribution to the pathogenesis of tumorigenesis in these patients.
Collapse
Affiliation(s)
- Liyun Qin
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Universitat Autònoma de Barcelona, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (L.Q.); (M.G.); (V.C.); (A.S.-F.); (J.T.); (M.A.)
| | - Maria Guitart
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Universitat Autònoma de Barcelona, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (L.Q.); (M.G.); (V.C.); (A.S.-F.); (J.T.); (M.A.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
| | - Víctor Curull
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Universitat Autònoma de Barcelona, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (L.Q.); (M.G.); (V.C.); (A.S.-F.); (J.T.); (M.A.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
| | - Albert Sánchez-Font
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Universitat Autònoma de Barcelona, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (L.Q.); (M.G.); (V.C.); (A.S.-F.); (J.T.); (M.A.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
| | - Xavier Duran
- Scientific and Technical Department, Hospital del Mar-IMIM, 08003 Barcelona, Spain;
| | - Jun Tang
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Universitat Autònoma de Barcelona, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (L.Q.); (M.G.); (V.C.); (A.S.-F.); (J.T.); (M.A.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
| | - Mireia Admetlló
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Universitat Autònoma de Barcelona, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (L.Q.); (M.G.); (V.C.); (A.S.-F.); (J.T.); (M.A.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
| | - Esther Barreiro
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Universitat Autònoma de Barcelona, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (L.Q.); (M.G.); (V.C.); (A.S.-F.); (J.T.); (M.A.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
- Correspondence: ; Tel.: +34-93-316-0385; Fax: +34-93-316-0410
| |
Collapse
|
43
|
Kinker GS, Vitiello GAF, Ferreira WAS, Chaves AS, Cordeiro de Lima VC, Medina TDS. B Cell Orchestration of Anti-tumor Immune Responses: A Matter of Cell Localization and Communication. Front Cell Dev Biol 2021; 9:678127. [PMID: 34164398 PMCID: PMC8215448 DOI: 10.3389/fcell.2021.678127] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/27/2021] [Indexed: 01/06/2023] Open
Abstract
The immune system plays a crucial role in cancer development either by fostering tumor growth or destroying tumor cells, which has open new avenues for cancer immunotherapy. It was only over the last decade that the role of B cells in controlling anti-tumor immune responses in the tumor milieu has begun to be appreciated. B and plasma cells can exert anti-tumor effects through antibody-dependent cell cytotoxicity (ADCC) and activation of the complement cascade, even though their effector functions extend beyond the classical humoral immunity. In tumor tissues, B cells can be found in lymphoid aggregates, known as tertiary lymphoid structures (TLSs), well-organized non-encapsulated structures composed of immune and stromal cells. These structures reflect a process of lymphoid neogenesis occurring in peripheral tissues upon long-lasting exposure to inflammatory signals. The TLS provides an area of intense B cell antigen presentation that can lead to optimal T cell activation and effector functions, as well as the generation of effector B cells, which can be further differentiated in either antibody-secreting plasma cells or memory B cells. Of clinical interest, the crosstalk between B cells and antigen-experienced and exhausted CD8+ T cells within mature TLS was recently associated with improved response to immune checkpoint blockade (ICB) in melanoma, sarcoma and lung cancer. Otherwise, B cells sparsely distributed in the tumor microenvironment or organized in immature TLSs were found to exert immune-regulatory functions, inhibiting anti-tumor immunity through the secretion of anti-inflammatory cytokines. Such phenotype might arise when B cells interact with malignant cells rather than T and dendritic cells. Differences in the spatial distribution likely underlie discrepancies between the role of B cells inferred from human samples or mouse models. Many fast-growing orthotopic tumors develop a malignant cell-rich bulk with reduced stroma and are devoid of TLSs, which highlights the importance of carefully selecting pre-clinical models. In summary, strategies that promote TLS formation in close proximity to tumor cells are likely to favor immunotherapy responses. Here, the cellular and molecular programs coordinating B cell development, activation and organization within TLSs will be reviewed, focusing on their translational relevance to cancer immunotherapy.
Collapse
Affiliation(s)
- Gabriela Sarti Kinker
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Glauco Akelinghton Freire Vitiello
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
- Department of Pathological Sciences, Londrina State University, Londrina, Brazil
| | - Wallax Augusto Silva Ferreira
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
- Laboratory of Tissue Culture and Cytogenetics, Environment Section (SAMAM), Evandro Chagas Institute, Ananindeua, Brazil
| | - Alexandre Silva Chaves
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
| | | | - Tiago da Silva Medina
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
- National Institute of Science and Technology in Oncogenomics and Therapeutic Innovation, São Paulo, Brazil
| |
Collapse
|
44
|
Curtis JL. Wouldn't you like to know: are tertiary lymphoid structures necessary for lung defence? Eur Respir J 2021; 57:57/4/2004352. [PMID: 33858851 DOI: 10.1183/13993003.04352-2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 12/08/2020] [Indexed: 11/05/2022]
Affiliation(s)
- Jeffrey L Curtis
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, MI, USA .,Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA.,Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
45
|
N J, J T, Sl N, Gt B. Tertiary lymphoid structures and B lymphocytes in cancer prognosis and response to immunotherapies. Oncoimmunology 2021; 10:1900508. [PMID: 33854820 PMCID: PMC8018489 DOI: 10.1080/2162402x.2021.1900508] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopic cellular aggregates that resemble secondary lymphoid organs in their composition and structural organization. In contrast to secondary lymphoid organs, TLS are not imprinted during embryogenesis but are formed in non-lymphoid tissues in response to local inflammation. TLS structures exhibiting a variable degree of maturation are found in solid tumors. They are composed of various immune cell types including dendritic cells and antigen-specific B and T lymphocytes, that together, actively drive the immune response against tumor development and progression. This review highlights the successive steps leading to tumor TLS formation and its association with clinical outcomes. We discuss the role played by tumor-infiltrating B lymphocytes and plasma cells, their prognostic value in solid tumors and immunotherapeutic responses and their potential for future targeting.
Collapse
Affiliation(s)
- Jacquelot N
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Tellier J
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Nutt Sl
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Belz Gt
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,The University of Queensland Diamantina Institute, the University of Queensland, Brisbane, Australia
| |
Collapse
|
46
|
Liu M, Zheng Q, Chen S, Liu J, Li S. FUT7 Promotes the Epithelial-Mesenchymal Transition and Immune Infiltration in Bladder Urothelial Carcinoma. J Inflamm Res 2021; 14:1069-1084. [PMID: 33790621 PMCID: PMC8007615 DOI: 10.2147/jir.s296597] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
Background Bladder urothelial carcinoma (BLCA) is one of the most frequently appearing, lethal and aggressive malignancies of the genitourinary system with growing morbidity and mortality, which affects human health seriously. Protein glycosylation, catalyzed by specific glycosyltransferase, has been found to be abnormal in several diseases, especially cancer. Fucosyltransferase VII (FUT7), one of the fucosyltransferases, was observed abnormally expressed in various cancers, however, the role of FUT7 in BLCA, and the association between its expression and clinical outcomes or immune infiltration remains unclear. Methodology FUT7 expression in BLCA was analyzed in Oncomine database, which was further confirmed with immunohistochemistry and ELISA. The prognostic value of FUT7 for BLCA was evaluated with PrognoScan database, and its genetic alteration was examined in cBioPortal database. The proliferation, migration, invasion and epithelial–mesenchymal transition (EMT) changes of bladder cancer cells after FUT7 siRNA or cDNA transfection were determined by CCK8, colony formation, transwell and Western blot, respectively. The correlation between FUT7 expression and immune infiltration levels was analyzed in TIMER and TISIDB databases, and the methylation level of FUT7 was detected in UALCAN database. Results The results showed that the expression of FUT7 was increased in BLCA, and patients with high FUT7 level were predicted to have lower overall survival and disease-specific survival rates, which were not influenced by FUT7 genetic alterations. Downregulation FUT7 inhibited the proliferation, migration, invasion and EMT of bladder cancer cells, whereas upregulation of FUT7 showed the opposite effects. We found that FUT7 was positively correlated with immune cell infiltration levels (CD8+ T cells, CD4+T cells, macrophage, neutrophil and dendritic cells), and also the expression of gene markers of immune cells. The negative correlation between FUT7 expression and FUT7 methylation level was observed, among which FUT7 expression was positively correlated with the abundance of 28 kinds of tumor-infiltrating lymphocytes (TILs), while FUT7 methylation level was negatively correlated with TILs. Conclusion Altogether, these findings suggested that FUT7 possessed the potential to serve as a detection biomarker or immunotherapeutic target for BLCA.
Collapse
Affiliation(s)
- Mulin Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, People's Republic of China
| | - Qin Zheng
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning Province, 116044, People's Republic of China
| | - Siyi Chen
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning Province, 116044, People's Republic of China
| | - Jiwei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, People's Republic of China
| | - Shijun Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, People's Republic of China
| |
Collapse
|
47
|
Tang J, Curull V, Wang X, Ampurdanés C, Duran X, Pijuan L, Rodríguez-Fuster A, Aguiló R, Yélamos J, Barreiro E. Increased PARP Activity and DNA Damage in NSCLC Patients: The Influence of COPD. Cancers (Basel) 2020; 12:E3333. [PMID: 33187221 PMCID: PMC7697659 DOI: 10.3390/cancers12113333] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
(1) Background: Lung cancer (LC) is a major leading cause of death worldwide. Poly (ADP-ribose) polymerase (PARP)-1 and PARP-2 are key players in cancer. We aimed to assess PARP-1 and PARP-2 expression and activity and DNA damage in tumors and non-tumor lungs from patients with/without chronic obstructive pulmonary disease (COPD). (2) Methods: Lung tumor and non-tumor specimens were obtained through video-assisted thoracoscopic surgery (VATS) in LC patients with/without underlying COPD (two groups of patients, n = 15/group). PARP-1 and PARP-2 expression (ELISA), PARP activity (PARP colorimetric assay kit) and DNA damage (immunohistochemistry) levels were identified in all samples. (3) Results: Both PARP-1 and PARP-2 expression levels were significantly lower in lung tumors (irrespective of COPD)compared to non-tumor specimens, while DNA damage and PARP activity levels significantly increased in lung tumors compared to non-tumor specimens only in LC-COPD patients. PARP-2 expression was positively correlated with smoking burden in LC-COPD patients. (4) Conclusions: In lung tumors of COPD patients, an overactivation of PARP enzyme was observed. A decline in PARP-1 and PARP-2 protein expression was seen in lung tumors irrespective of COPD. Other phenotypic features (airway obstruction) beyond cancer may account for the increase in PARP activity seen in the tumors of patients with underlying COPD.
Collapse
Affiliation(s)
- Jun Tang
- Pulmonology Department, Lung Cancer and Muscle Research Group, Hospital del Mar-IMIM, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Medical School, Universitat Autònoma de Barcelona, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (J.T.); (V.C.); (X.W.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
| | - Víctor Curull
- Pulmonology Department, Lung Cancer and Muscle Research Group, Hospital del Mar-IMIM, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Medical School, Universitat Autònoma de Barcelona, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (J.T.); (V.C.); (X.W.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
| | - Xuejie Wang
- Pulmonology Department, Lung Cancer and Muscle Research Group, Hospital del Mar-IMIM, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Medical School, Universitat Autònoma de Barcelona, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (J.T.); (V.C.); (X.W.)
| | - Coral Ampurdanés
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM)-Hospital del Mar, 08003 Barcelona, Spain; (C.A.); (J.Y.)
| | - Xavier Duran
- Scientific, Statistics and Technical Department, Hospital del Mar-IMIM, Parc de Salut Mar, 08003 Barcelona, Spain;
| | - Lara Pijuan
- Pathology Department, Hospital del Mar-IMIM, Parc de Salut Mar, 08003 Barcelona, Spain;
| | - Alberto Rodríguez-Fuster
- Thoracic Surgery Department, Hospital del Mar-IMIM, Parc de Salut Mar, 08003 Barcelona, Spain; (A.R.-F.); (R.A.)
| | - Rafael Aguiló
- Thoracic Surgery Department, Hospital del Mar-IMIM, Parc de Salut Mar, 08003 Barcelona, Spain; (A.R.-F.); (R.A.)
| | - José Yélamos
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM)-Hospital del Mar, 08003 Barcelona, Spain; (C.A.); (J.Y.)
- Pathology Department, Hospital del Mar-IMIM, Parc de Salut Mar, 08003 Barcelona, Spain;
| | - Esther Barreiro
- Pulmonology Department, Lung Cancer and Muscle Research Group, Hospital del Mar-IMIM, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Medical School, Universitat Autònoma de Barcelona, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (J.T.); (V.C.); (X.W.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
| |
Collapse
|