1
|
Elfving H, Yu H, Fessehatsion KK, Brunnström H, Botling J, Gulyas M, Backman M, Lindberg A, Strell C, Micke P. Spatial distribution of tertiary lymphoid structures in the molecular and clinical context of non-small cell lung cancer. Cell Oncol (Dordr) 2025; 48:801-813. [PMID: 40029549 DOI: 10.1007/s13402-025-01052-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 03/05/2025] Open
Abstract
INTRODUCTION Tertiary lymphoid structures (TLS) are lymphocyte aggregates resembling secondary lymphoid organs and are pivotal in cancer immunity. The ambiguous morphological definition of TLS makes it challenging to ascertain their clinical impact on patient survival and response to immunotherapy. OBJECTIVES This study aimed to characterize TLS in hematoxylin-eosin tissue sections from lung cancer patients, assessing their occurrence in relation to the local immune environment, mutational background, and patient outcome. METHODS Two pathologists evaluated one whole tissue section from resection specimens of 680 NSCLC patients. TLS were spatially quantified within the tumor area or periphery and further categorized based on the presence of germinal centers (mature TLS). Metrics were integrated with immune cell counts, genomic and transcriptomic data, and correlated with clinical parameters. RESULTS TLS were present in 86% of 536 evaluable cases, predominantly in the tumor periphery, with a median of eight TLS per case. Mature TLS were found in 24% of cases. TLS presence correlated positively with increased plasma cell (CD138+) and lymphocytic cell (CD3+, CD8+, FOXP3+) infiltration. Tumors with higher tumor mutational burden exhibited higher numbers of peripheral TLS. The overall TLS quantity was independently associated with improved patient survival, irrespective of TLS maturation status. This prognostic association held true for peripheral TLS but not for tumor TLS. CONCLUSION TLS in NSCLC is common and their correlation with a specific immune phenotype suggests biological relevance in the local immune reaction. The prognostic significance of this scoring system on routine hematoxylin-eosin sections has the potential to augment diagnostic algorithms for NSCLC patients.
Collapse
Affiliation(s)
- Hedvig Elfving
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden.
| | - Hui Yu
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | | | - Hans Brunnström
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Johan Botling
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Miklos Gulyas
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Max Backman
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Amanda Lindberg
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Carina Strell
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Patrick Micke
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| |
Collapse
|
2
|
Zhou X, Cheng S, Chen Z, Zhang J, Wang J, Li Q, Zhou X. Inhibiting HnRNP L-mediated alternative splicing of EIF4G1 counteracts immune checkpoint blockade resistance in Castration-resistant prostate Cancer. Neoplasia 2025; 60:101109. [PMID: 39724754 PMCID: PMC11731738 DOI: 10.1016/j.neo.2024.101109] [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: 06/28/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Immunotherapy with checkpoint inhibitors produced significant clinical responses in a subset of cancer patients who were resistant to prior therapies. However, Castration-resistant prostate cancer (CRPC) is seriously lack of T cell infiltration, which greatly limits the clinical application of immunotherapy, but the mechanism is unclear. In the present study, in silico analyses and experimental data show that HnRNP L was significantly negatively correlated with CD4+ and CD8+ T cells infiltration in patients; besides, we found deficiency of HnRNP L recruites CD4+ and CD8+ T cells infiltration and impairs tumorigenesis. Mechanically, HnRNP L enhanced the translation of c-Myc and then promoted CXCL8 secretion via alternative splicing of EIF4G1. In vivo, inhibition of EIF4G1 by the inhibitor, SBI-0640756, attenuated HnRNP l-induced tumor progression and immunosuppressive activity. And most of all, therapeutic synergy between HnRNP L knockdown and Anti-PD-1 could significantly suppress xenograft prostate cancer growth. In summary, this study revealled the molecular mechanism of HnRNP L regulating the immune infiltration, which provides a new theoretical basis for overcoming the limitation of immunotherapy for CRPC.
Collapse
MESH Headings
- Male
- Humans
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/immunology
- Animals
- Mice
- Alternative Splicing
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Drug Resistance, Neoplasm/genetics
- Eukaryotic Initiation Factor-4G/genetics
- Eukaryotic Initiation Factor-4G/metabolism
- Heterogeneous-Nuclear Ribonucleoprotein L/genetics
- Heterogeneous-Nuclear Ribonucleoprotein L/metabolism
- Heterogeneous-Nuclear Ribonucleoprotein L/antagonists & inhibitors
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Gene Expression Regulation, Neoplastic
- Disease Models, Animal
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
Collapse
Affiliation(s)
- Xumin Zhou
- General Surgery Center Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China
| | - Shilong Cheng
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China
| | - Zhongjie Chen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China
| | - Jinming Zhang
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Jiaqi Wang
- Department of Spinal Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China
| | - Qiang Li
- General Surgery Center Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China.
| | - Xumin Zhou
- General Surgery Center Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China.
| |
Collapse
|
3
|
Yue S, Zhang Y, Zhang W. Recent Advances in Immunotherapy for Advanced Biliary Tract Cancer. Curr Treat Options Oncol 2024; 25:1089-1111. [PMID: 39066855 PMCID: PMC11329538 DOI: 10.1007/s11864-024-01243-y] [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] [Accepted: 06/21/2024] [Indexed: 07/30/2024]
Abstract
OPINION STATEMENT Biliary tract cancer (BTC) is a heterogeneous group of aggressive malignancies that arise from the epithelium of the biliary tract. Most patients present with locally advanced or metastatic disease at the time of diagnosis. For patients with unresectable BTC, the survival advantage provided by systemic chemotherapy was limited. Over the last decade, immunotherapy has significantly improved the therapeutic landscape of solid tumors. There is an increasing number of studies evaluating the application of immunotherapy in BTC, including immune checkpoint inhibitors (ICIs), cancer vaccines and adoptive cell therapy. The limited response to ICIs monotherapy in unselected patients prompted investigators to explore different combination therapy strategies. Early clinical trials of therapeutic cancer vaccination and adoptive cell therapy have shown encouraging clinical results. However, there still has been a long way to go via validation of therapeutic efficacy and exploration of strategies to increase the efficacy. Identifying biomarkers that predict the response to immunotherapy will allow a more accurate selection of candidates. This review will provide an up-to-date overview of the current clinical data on the role of immunotherapy, summarize the promising biomarkers predictive of the response to ICIs and discuss the perspective for future research direction of immunotherapy in advanced BTC.
Collapse
Affiliation(s)
- Shiwei Yue
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, China
- Hubei Key Laboratory of Hepato‑Pancreatic‑Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, 1095 Jiefang Avenue, 430030, Wuhan, China
| | - Yunpu Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, China
- Hubei Key Laboratory of Hepato‑Pancreatic‑Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, 1095 Jiefang Avenue, 430030, Wuhan, China
| | - Wei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, China.
- Hubei Key Laboratory of Hepato‑Pancreatic‑Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, 1095 Jiefang Avenue, 430030, Wuhan, China.
| |
Collapse
|
4
|
Li X, Eastham J, Giltnane JM, Zou W, Zijlstra A, Tabatsky E, Banchereau R, Chang CW, Nabet BY, Patil NS, Molinero L, Chui S, Harryman M, Lau S, Rangell L, Waumans Y, Kockx M, Orlova D, Koeppen H. Automated tumor immunophenotyping predicts clinical benefit from anti-PD-L1 immunotherapy. J Pathol 2024; 263:190-202. [PMID: 38525811 DOI: 10.1002/path.6274] [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: 07/01/2023] [Revised: 12/22/2023] [Accepted: 02/14/2024] [Indexed: 03/26/2024]
Abstract
Cancer immunotherapy has transformed the clinical approach to patients with malignancies, as profound benefits can be seen in a subset of patients. To identify this subset, biomarker analyses increasingly focus on phenotypic and functional evaluation of the tumor microenvironment to determine if density, spatial distribution, and cellular composition of immune cell infiltrates can provide prognostic and/or predictive information. Attempts have been made to develop standardized methods to evaluate immune infiltrates in the routine assessment of certain tumor types; however, broad adoption of this approach in clinical decision-making is still missing. We developed approaches to categorize solid tumors into 'desert', 'excluded', and 'inflamed' types according to the spatial distribution of CD8+ immune effector cells to determine the prognostic and/or predictive implications of such labels. To overcome the limitations of this subjective approach, we incrementally developed four automated analysis pipelines of increasing granularity and complexity for density and pattern assessment of immune effector cells. We show that categorization based on 'manual' observation is predictive for clinical benefit from anti-programmed death ligand 1 therapy in two large cohorts of patients with non-small cell lung cancer or triple-negative breast cancer. For the automated analysis we demonstrate that a combined approach outperforms individual pipelines and successfully relates spatial features to pathologist-based readouts and the patient's response to therapy. Our findings suggest that tumor immunophenotype generated by automated analysis pipelines should be evaluated further as potential predictive biomarkers for cancer immunotherapy. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Xiao Li
- Genentech, South San Francisco, CA, USA
| | | | | | - Wei Zou
- Genentech, South San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | - Shari Lau
- Genentech, South San Francisco, CA, USA
| | | | | | | | | | | |
Collapse
|
5
|
Zhang C, Fei Y, Wang H, Hu S, Liu C, Hu R, Du Q. CAFs orchestrates tumor immune microenvironment—A new target in cancer therapy? Front Pharmacol 2023; 14:1113378. [PMID: 37007004 PMCID: PMC10064291 DOI: 10.3389/fphar.2023.1113378] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/23/2023] [Indexed: 03/15/2023] Open
Abstract
Cancer immunotherapy has opened a new landscape in cancer treatment, however, the poor specificity and resistance of most targeted therapeutics have limited their therapeutic efficacy. In recent years, the role of CAFs in immune regulation has been increasingly noted as more evidence has been uncovered regarding the link between cancer-associated fibroblasts (CAFs) and the evolutionary process of tumor progression. CAFs interact with immune cells to shape the tumor immune microenvironment (TIME) that favors malignant tumor progression, a crosstalk process that leads to the failure of cancer immunotherapies. In this review, we outline recent advances in the immunosuppressive function of CAFs, highlight the mechanisms of CAFs-immune cell interactions, and discuss current CAF-targeted therapeutic strategies for future study.
Collapse
Affiliation(s)
- Chunxue Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuxiang Fei
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hui Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Sheng Hu
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Chao Liu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Qianming Du, ; Rong Hu, ; Chao Liu,
| | - Rong Hu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Jiangsu Nanjing, China
- *Correspondence: Qianming Du, ; Rong Hu, ; Chao Liu,
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Qianming Du, ; Rong Hu, ; Chao Liu,
| |
Collapse
|
6
|
Peng Y, Li Z, Fu Y, Pan Y, Zeng Y, Liu J, Xiao C, Zhang Y, Su Y, Li G, Wu F. Progress and perspectives of perioperative immunotherapy in non-small cell lung cancer. Front Oncol 2023; 13:1011810. [PMID: 36761954 PMCID: PMC9905802 DOI: 10.3389/fonc.2023.1011810] [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: 08/04/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023] Open
Abstract
Lung cancer is one of the leading causes of cancer-related death. Lung cancer mortality has decreased over the past decade, which is partly attributed to improved treatments. Curative surgery for patients with early-stage lung cancer is the standard of care, but not all surgical treatments have a good prognosis. Adjuvant and neoadjuvant chemotherapy are used to improve the prognosis of patients with resectable lung cancer. Immunotherapy, an epoch-defining treatment, has improved curative effects, prognosis, and tolerability compared with traditional and ordinary cytotoxic chemotherapy, providing new hope for patients with non-small cell lung cancer (NSCLC). Immunotherapy-related clinical trials have reported encouraging clinical outcomes in their exploration of different types of perioperative immunotherapy, from neoadjuvant immune checkpoint inhibitor (ICI) monotherapy, neoadjuvant immune-combination therapy (chemoimmunotherapy, immunotherapy plus antiangiogenic therapy, immunotherapy plus radiotherapy, or concurrent chemoradiotherapy), adjuvant immunotherapy, and neoadjuvant combined adjuvant immunotherapy. Phase 3 studies such as IMpower 010 and CheckMate 816 reported survival benefits of perioperative immunotherapy for operable patients. This review summarizes up-to-date clinical studies and analyzes the efficiency and feasibility of different neoadjuvant therapies and biomarkers to identify optimal types of perioperative immunotherapy for NSCLC.
Collapse
Affiliation(s)
- Yurong Peng
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhuo Li
- The Ophthalmologic Center of the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yucheng Fu
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yue Pan
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yue Zeng
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junqi Liu
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chaoyue Xiao
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yingzhe Zhang
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yahui Su
- XiangYa School of Public Health, Central South University, Changsha, Hunan, China
| | - Guoqing Li
- XiangYa School of Public Health, Central South University, Changsha, Hunan, China
| | - Fang Wu
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
Toledo B, Picon-Ruiz M, Marchal JA, Perán M. Dual Role of Fibroblasts Educated by Tumour in Cancer Behavior and Therapeutic Perspectives. Int J Mol Sci 2022; 23:15576. [PMID: 36555218 PMCID: PMC9778751 DOI: 10.3390/ijms232415576] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Tumours are complex systems with dynamic interactions between tumour cells, non-tumour cells, and extracellular components that comprise the tumour microenvironment (TME). The majority of TME's cells are cancer-associated fibroblasts (CAFs), which are crucial in extracellular matrix (ECM) construction, tumour metabolism, immunology, adaptive chemoresistance, and tumour cell motility. CAF subtypes have been identified based on the expression of protein markers. CAFs may act as promoters or suppressors in tumour cells depending on a variety of factors, including cancer stage. Indeed, CAFs have been shown to promote tumour growth, survival and spread, and secretome changes, but they can also slow tumourigenesis at an early stage through mechanisms that are still poorly understood. Stromal-cancer interactions are governed by a variety of soluble factors that determine the outcome of the tumourigenic process. Cancer cells release factors that enhance the ability of fibroblasts to secrete multiple tumour-promoting chemokines, acting on malignant cells to promote proliferation, migration, and invasion. This crosstalk between CAFs and tumour cells has given new prominence to the stromal cells, from being considered as mere physical support to becoming key players in the tumour process. Here, we focus on the concept of cancer as a non-healing wound and the relevance of chronic inflammation to tumour initiation. In addition, we review CAFs heterogeneous origins and markers together with the potential therapeutic implications of CAFs "re-education" and/or targeting tumour progression inhibition.
Collapse
Affiliation(s)
- Belén Toledo
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
| | - Manuel Picon-Ruiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| |
Collapse
|
8
|
Lin M, Huang Z, Chen Y, Xiao H, Wang T. Lung cancer patients with chronic obstructive pulmonary disease benefit from anti-PD-1/PD-L1 therapy. Front Immunol 2022; 13:1038715. [PMID: 36532019 PMCID: PMC9751394 DOI: 10.3389/fimmu.2022.1038715] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/17/2022] [Indexed: 12/04/2022] Open
Abstract
Lung cancer (LC) and chronic obstructive pulmonary disease (COPD) are two of the most fatal respiratory diseases, seriously threatening human health and imposing a heavy burden on families and society. Although COPD is a significant independent risk factor for LC, it is still unclear how COPD affects the prognosis of LC patients, especially when LC patients with COPD receive immunotherapy. With the development of immune checkpoint inhibition (ICI) therapy, an increasing number of inhibitors of programmed cell death-1 (PD-1) and PD-1 ligand (PD-L1) have been applied to the treatment of LC. Recent studies suggest that LC patients with COPD may benefit more from immunotherapy. In this review, we systematically summarized the outcomes of LC patients with COPD after anti-PD-1/PD-L1 treatment and discussed the tumor immune microenvironment (TIME) regulated by COPD in LC immunotherapy, which provides novel insights for the clinical treatment of LC patients with COPD.
Collapse
Affiliation(s)
- Mao Lin
- Department of Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zongyao Huang
- Department of Pathology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yingfu Chen
- Department of Pharmacy, Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Hongtao Xiao
- Department of Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ting Wang
- Department of Clinical Research, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China,*Correspondence: Ting Wang,
| |
Collapse
|
9
|
Fan L, Wang X, Chang Q, Wang Y, Yang W, Liu L. IL2RA is a prognostic indicator and correlated with immune characteristics of pancreatic ductal adenocarcinoma. Medicine (Baltimore) 2022; 101:e30966. [PMID: 36281157 PMCID: PMC9592409 DOI: 10.1097/md.0000000000030966] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive and incurable cancer with a dismal prognosis. In this study, we aimed to explore potential predictors for the prognosis and immunological characteristics of PDAC. Estimation of stromal and immune cells in malignant tumors, using expression data (ESTIMATE) method was applied to calculate the immune and stromal scores of 206 PDAC samples from GSE71729. R package of "limma" was utilized to identify differentially expressed genes (DEGs). Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichment analyses were conducted for functional exploration. Protein-protein interaction (PPI) network and Univariate Cox analysis were conducted to select key prognostic genes of PDAC. Gene set enrichment analysis (GSEA) was applied to investigate the roles of IL2RA in PDAC. Single sample GSEA (ssGSEA) was performed to evaluate the immunological characteristics of PDAC samples. Wilcoxon rank sum test was conducted to compare the difference of immunological characteristics of PDAC samples between low IL2RA and high IL2RA. Spearman correlation analysis was used to explore the correlations of IL2RA expression and immune checkpoint genes. A total of 747 DEGs were identified between low and high immune/stromal groups. Functional exploration revealed upregulated DEGs were associated with immune-related activities, whereas downregulated DEGs were involved in inflammatory-related activities. IL2RA was selected as the critical gene by overlapping the hub genes in PPI network and prognostic genes. Significantly, IL2RA expression was significantly elevated in PDAC and patients with higher IL2RA expression had worse prognoses. The immunological and oncogenic roles of IL2RA in PDAC were evidenced by GSEA. Furthermore, PDAC samples with high IL2RA expression exhibited increased immune infiltration and better immunotherapy responses. IL2RA expression was positively correlated with PDCD1, CD274, CTLA4, IDO1, TDO2, and TIGT. Higher expression of IL2RA predicts worse survival outcomes and increased immune infiltration in PDAC. PDAC patients with high IL2RA expression might potentially benefit from immunotherapy.
Collapse
Affiliation(s)
- Liwen Fan
- Department of Radiotherapy, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xinyu Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qing Chang
- Department of Radiotherapy, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yue Wang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Wenjie Yang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Linlin Liu
- Department of Radiotherapy, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
- *Correspondence: Linlin Liu, Department of Radiotherapy, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China (e-mail: )
| |
Collapse
|
10
|
Tian W, Yang Y, Qin Q, Zhang L, Wang Z, Su L, Zeng L, Chen H, Hu L, Hong J, Huang Y, Zhang Q, Zhao H. Vimentin and tumor-stroma ratio for neoadjuvant chemoradiotherapy response prediction in locally advanced rectal cancer. Cancer Sci 2022; 114:619-629. [PMID: 36221784 PMCID: PMC9899599 DOI: 10.1111/cas.15610] [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: 06/16/2022] [Revised: 09/11/2022] [Accepted: 09/22/2022] [Indexed: 01/07/2023] Open
Abstract
Vimentin expression in tumor tissues and the tumor-stroma ratio (TSR) have been demonstrated as strong prognostic factors for cancer patients, but whether they are predictive markers of neoadjuvant chemoradiotherapy (nCRT) outcome in locally advanced rectal cancer (LARC) patients is poorly understood. This study aimed to explore the predictive significance of vimentin and TSR combined for nCRT response in LARC patients. Imaging mass cytometry (IMC) was performed to determine the association of vimentin and TSR with nCRT response in six LARC patients [three achieved pathological complete response (pCR), three did not]. Immunohistochemistry (IHC) for vimentin and TSR on biopsy tissues before nCRT and logistic regression analysis were performed to further evaluate their predictive value for treatment responses in a larger patient cohort. A trend of decreased vimentin expression and increased TSR in the pCR group was revealed by IMC. In the validation group, vimentin [odds ratio (OR) 0.260, 95% confidence interval (CI) 0.102-0.602, p = 0.002] and TSR (OR 4.971, 95% CI 1.933-15.431, p = 0.002) were associated with pCR by univariate analysis. Patients in the vimentin-low/TSR-low or vimentin-high/TSR-high (OR 5.211, 95% CI 1.248-35.582, p = 0.042) and vimentin-low/TSR-high groups (OR 11.846, 95% CI 3.197-77.079, p = 0.001) had significantly higher odds of pCR. By multivariate analysis, only the combination of vimentin and TSR was an independent predictor for nCRT response (OR 9.324, 95% CI 2.290-63.623, p = 0.006). Our study suggested that the combined assessment of vimentin and TSR can provide additive significance and may be a promising indicator of nCRT response in LARC patients.
Collapse
Affiliation(s)
- Wenjing Tian
- The Cancer Center of the Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated HospitalSun Yat‐sen UniversityZhuhaiChina
| | - Yuqin Yang
- The Cancer Center of the Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina,Department of Pathology, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina,Department of Pathology, School of Basic Medical ScienceSouthern Medical UniversityGuangzhouChina
| | - Qi Qin
- Department of Medical OncologyThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Liguo Zhang
- Department of Thyroid & Breast Surgery, The Fifth Affiliated HospitalSun Yat‐sen UniversityZhuhaiChina
| | - Zheyan Wang
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated HospitalSun Yat‐sen UniversityZhuhaiChina
| | - Liqian Su
- Precision Medicine Center of Harbin Medical University Cancer HospitalHarbinChina
| | - Lirong Zeng
- The Cancer Center of the Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Hui Chen
- The Cancer Center of the Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Lingzhi Hu
- The Cancer Center of the Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Jiawei Hong
- Department of Thyroid & Breast Surgery, The Fifth Affiliated HospitalSun Yat‐sen UniversityZhuhaiChina
| | - Ying Huang
- The Cancer Center of the Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina,Department of Pathology, School of Basic Medical ScienceSouthern Medical UniversityGuangzhouChina
| | - Hong Zhao
- The Cancer Center of the Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated HospitalSun Yat‐sen UniversityZhuhaiChina
| |
Collapse
|
11
|
Immunotherapy for Aggressive and Metastatic Pituitary Neuroendocrine Tumors (PitNETs): State-of-the Art. Cancers (Basel) 2022; 14:cancers14174093. [PMID: 36077631 PMCID: PMC9454884 DOI: 10.3390/cancers14174093] [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: 07/21/2022] [Revised: 08/20/2022] [Accepted: 08/21/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Aggressive and metastatic PitNETs are challenging conditions. Immune checkpoint inhibitors (ICIs) are currently considered in cases resistant to temozolomide (TMZ). However, clinical experience is essentially limited to case reports, with variable outcomes. Material and Methods: The effects of ICIs on 12 aggressive/metastatic PitNETs from the literature were reviewed and analyzed according to tumor characteristics, with the additional description of a silent-Pit1 metastatic tumor responding to pembrolizumab. Results: Most cases were metastatic (10/13: 6 corticotroph, 3 lactotroph, 1 silent Pit1); 3 were aggressive (2 corticotroph, 1 lactotroph). ICIS was used either as monotherapy or in combination. At last follow-up on ICI, a complete response (CR) was present in 3 cases and a partial response (PR) in 2 cases (4/5 metastatic). One sustained stable disease (SD) was reported. Progressive disease (PD) was observed in 7 cases, 3 of them after initial SD (n = 1) or PR (n = 3), with 2 reported deaths. PDL1 expression was studied in 10 cases and was high (>95%) in 2 Pit1-derived metastatic PitNETs (1 CR and 1 remarkable PR) but absent/low (<1%) in the remaining cases (including 1 CP and 2 PR). Elevated tumor mutation burden could be informative in corticotroph PitNETs, especially in mismatch repair-deficient tumors. Conclusion: Significant benefits from ICIs were documented in about half of TMZ-resistant PitNETS. High PDL1 expression was associated with remarkable responses but may be dispensable. Based on their acceptable tolerance and awaiting recognized predictors of response, ICIs may be considered a valuable option for such patients.
Collapse
|
12
|
Jiang D, Song Q, Wei X, Yu Z, Liu Y, Wang H, Wang X, Huang J, Su J, Hong Y, Xu Y, Xu C, Hou Y. PMS2 Expression With Combination of PD-L1 and TILs for Predicting Survival of Esophageal Squamous Cell Carcinoma. Front Oncol 2022; 12:897527. [PMID: 35865481 PMCID: PMC9294642 DOI: 10.3389/fonc.2022.897527] [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: 03/16/2022] [Accepted: 06/02/2022] [Indexed: 11/17/2022] Open
Abstract
Background DNA mismatch repair (MMR) deficiency (dMMR) has been recognized as an important biomarker for immunotherapy in esophageal squamous cell carcinoma (ESCC), along with programmed death ligand 1 (PD-L1) expression and/or tumor-infiltrated lymphocytes (TILs). However, in ESCC, MMR protein assessment has not been well studied at present. Methods A total of 484 ESCC tissues treated between 2007 and 2010, in our hospital, were enrolled. Immunohistochemical expression of MLH1, MSH2, MSH6, PMS2, and PD-L1 on tissue microarray specimens and clinicopathological features, including TILs, were analyzed retrospectively. Results Out of the 484 studied cases, loss of MLH1, MSH2, MSH6, and PMS2 expression were found in 6.8%, 2.1%, 8.7%, and 4.8% patients, respectively. dMMR was found in 65 patients, 37 cases involved in one MMR protein, 17 cases involved in two proteins, 7 cases involved in three proteins, and 4 cases involved in four proteins. There was no significant survival difference between pMMR (MMR-proficient) and dMMR patients (P>0.05). However, 224 patients with low PMS2 expression had better DFS and OS than 260 patients with high PMS2 expression (P=0.006 for DFS and 0.008 for OS), which was identified as an independent prognostic factor in multivariate analyses. Positive PD-L1 expression was detected in 341 (70.5%) samples. In stage I-II disease, patients with PD-L1 expression had better DFS and OS than those without PD-L1 expression(P<0.05), which was not found in stage III-IV disease. With the ITWG system, 40.1% of cases were classified as high TILs. Patients in the high-TILs group tended to have better DFS (P=0.055) and OS (P=0.070) than those in the low-TILs group and the differences were statistically significant in pMMR, high MSH6, or PMS2 expression cases (P<0.05). Also, high PMS2 expression patients with both PD-L1 expression and high TILs, had similar DFS and OS compared with low PMS2 expression patients (P>0.05), which were much better than other high PMS2 expression patients. Conclusion The expression level of MMR proteins could also be used as a prognostic factor in ESCC and PMS2 expression outperformed other MMR proteins for predicting survival. The combination of PD-L1 expression and TILs may lead to more efficient risk stratification of ESCC.
Collapse
Affiliation(s)
- Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Qi Song
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaojun Wei
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zixiang Yu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yufeng Liu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haixing Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xingxing Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jieakesu Su
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Hong
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifan Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Yingyong Hou, ; Chen Xu,
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- *Correspondence: Yingyong Hou, ; Chen Xu,
| |
Collapse
|
13
|
Saito Y, Fujiwara Y, Shinchi Y, Mito R, Miura Y, Yamaguchi T, Ikeda K, Urakami S, Nakashima Y, Sakagami T, Suzuki M, Tabata Y, Komohara Y. Classification of PD-L1 expression in various cancers and macrophages based on immunohistocytological analysis. Cancer Sci 2022; 113:3255-3266. [PMID: 35633190 PMCID: PMC9459416 DOI: 10.1111/cas.15442] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 03/23/2022] [Accepted: 04/20/2022] [Indexed: 12/01/2022] Open
Abstract
Programmed death (PD)‐1/PD‐ligand 1 (PD‐L1) antibodies have shown an intense clinical effect in some patients with PD‐L1+ tumors, and their applications have rapidly expanded to various cancer types with or without the application of new companion diagnostics (CDx) with a lower cutoff value and inclusion of macrophage evaluation. However, the pathological background explaining the difference in the cutoff value remains unknown. To address this, we evaluated tissue array samples from 231 patients with lung adenocarcinoma, 186 with lung squamous cell carcinoma, and 38 with renal cell carcinoma (RCC) who were not receiving PD‐1/PD‐L1 antibodies to investigate the relationship between PD‐L1 expression on tumor cells and CD8+ T‐cell infiltration in tumor tissues. PD‐L1 expression in RCC was clearly lower than that in non–small‐cell lung cancer (NSCLC) tissue, whereas CD8+ T‐cell infiltration was low in all cancers. We next analyzed PD‐L1 expression by interferon (α, β, and γ) and LPS stimulation in both macrophages and 41 cancer cell lines derived from various organs and histological types. The PD‐L1 expression patterns were classified into three types, which differed depending on each organ or tissue type. Interestingly, NSCLC cell lines showed highly diverse PD‐L1 expression patterns compared with RCC cell lines. Conversely, PD‐L1 expression was stronger and more prolonged in macrophages than in typical cell lines. Here, we revealed the diversity of the PD‐L1 expression patterns in tumor cells and macrophages, demonstrating the pathological and cytological significance of the transition of cutoff values in PD‐L1 CDx for PD‐1/PD‐L1 antibody administration.
Collapse
Affiliation(s)
- Yoichi Saito
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Bioengineering, Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yusuke Shinchi
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,Department of Thoracic Surgery, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Remi Mito
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,Department of Respiratory Medicine, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuji Miura
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,Department of Medical Oncology, Toranomon Hospital, Tokyo, Japan
| | - Tomoya Yamaguchi
- Department of Cancer Biology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Koei Ikeda
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Yuta Nakashima
- Laboratory of Bioengineering, Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Takuro Sakagami
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Makoto Suzuki
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
14
|
Wang X, Wang J, Zhao J, Wang H, Chen J, Wu J. HMGA2 facilitates colorectal cancer progression via STAT3-mediated tumor-associated macrophage recruitment. Theranostics 2022; 12:963-975. [PMID: 34976223 PMCID: PMC8692921 DOI: 10.7150/thno.65411] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/20/2021] [Indexed: 12/17/2022] Open
Abstract
Rationale: Tumor-associated macrophages (TAMs), generally displaying the pro-tumor M2-like phenotype, strongly influence the progression of colorectal cancer (CRC) via their immunosuppressive activities. The high-mobility gene group A2 (HMGA2), an oncoprotein, is aberrantly overexpressed in CRC cells. However, the mechanisms by which tumor-derived HMGA2 modulates tumor microenvironment in CRC remain poorly understood. Methods:In vivo subcutaneous tumor xenograft model, azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced tumor mouse model and in vitro co-culture assays were used to investigate the Hmga2 role in TAM recruitment and polarization. Luciferase and chromatin immunoprecipitation (ChIP) assays were applied to examine the mechanism of HMGA2-mediated transcriptional regulation of signal transducer and activator of transcription 3 (STAT3). The CD68 correlation with patient outcome was analyzed in 167 human CRC tissues. Results: We found that HMGA2 in cancer cells promoted macrophage recruitment and M2 polarization in vitro and in vivo. HMGA2 directly bound to the STAT3 promoter to activate its transcription and subsequently induced CCL2 secretion, thus promoting macrophage recruitment. Our results from human CRC specimens also revealed a strong positive association between HMGA2 expression in tumor cells and CD68 expression in the stroma. We further showed that patients with an elevated CD68 expression had an unfavorable overall survival in all of the patients or in the subgroup with negative distant metastasis. Conclusion: Our work uncovers new insight into the link between the HMGA2/STAT3/CCL2 axis and macrophage recruitment in CRC. These findings provide a novel therapeutic option for targeting the HMGA2/STAT3/CCL2 axis in CRC.
Collapse
|
15
|
Borroni EM, Grizzi F. Cancer Immunoediting and beyond in 2021. Int J Mol Sci 2021; 22:13275. [PMID: 34948072 PMCID: PMC8703961 DOI: 10.3390/ijms222413275] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023] Open
Abstract
Human cancer has been depicted as a non-linear dynamic system that is discontinuous in space and time, but progresses through different sequential states (Figure 1) [...].
Collapse
Affiliation(s)
- Elena Monica Borroni
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20133 Milan, Italy;
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Fabio Grizzi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| |
Collapse
|
16
|
Backman M, La Fleur L, Kurppa P, Djureinovic D, Elfving H, Brunnström H, Mattsson JSM, Lindberg A, Pontén V, Eltahir M, Mangsbo S, Gulyas M, Isaksson J, Jirström K, Kärre K, Leandersson K, Mezheyeuski A, Pontén F, Strell C, Lindskog C, Botling J, Micke P. Infiltration of NK and plasma cells is associated with a distinct immune subset in non-small cell lung cancer. J Pathol 2021; 255:243-256. [PMID: 34339045 DOI: 10.1002/path.5772] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/13/2021] [Accepted: 07/28/2021] [Indexed: 12/23/2022]
Abstract
Immune cells of the tumor microenvironment are central but erratic targets for immunotherapy. The aim of this study was to characterize novel patterns of immune cell infiltration in non-small cell lung cancer (NSCLC) in relation to its molecular and clinicopathologic characteristics. Lymphocytes (CD3+, CD4+, CD8+, CD20+, FOXP3+, CD45RO+), macrophages (CD163+), plasma cells (CD138+), NK cells (NKp46+), PD1+, and PD-L1+ were annotated on a tissue microarray including 357 NSCLC cases. Somatic mutations were analyzed by targeted sequencing for 82 genes and a tumor mutational load score was estimated. Transcriptomic immune patterns were established in 197 patients based on RNA sequencing data. The immune cell infiltration was variable and showed only poor association with specific mutations. The previously defined immune phenotypic patterns, desert, inflamed, and immune excluded, comprised 30, 13, and 57% of cases, respectively. Notably, mRNA immune activation and high estimated tumor mutational load were unique only for the inflamed pattern. However, in the unsupervised cluster analysis, including all immune cell markers, these conceptual patterns were only weakly reproduced. Instead, four immune classes were identified: (1) high immune cell infiltration, (2) high immune cell infiltration with abundance of CD20+ B cells, (3) low immune cell infiltration, and (4) a phenotype with an imprint of plasma cells and NK cells. This latter class was linked to better survival despite exhibiting low expression of immune response-related genes (e.g. CXCL9, GZMB, INFG, CTLA4). This compartment-specific immune cell analysis in the context of the molecular and clinical background of NSCLC reveals two previously unrecognized immune classes. A refined immune classification, including traits of the humoral and innate immune response, is important to define the immunogenic potency of NSCLC in the era of immunotherapy. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Max Backman
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Linnéa La Fleur
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Pinja Kurppa
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Dijana Djureinovic
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hedvig Elfving
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hans Brunnström
- Division of Pathology, Lund University, Skåne University Hospital, Lund, Sweden
| | | | - Amanda Lindberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Victor Pontén
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Mohamed Eltahir
- Department of Pharmaceutical Bioscience, Uppsala University, Uppsala, Sweden
| | - Sara Mangsbo
- Department of Pharmaceutical Bioscience, Uppsala University, Uppsala, Sweden
| | - Miklos Gulyas
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Johan Isaksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Respiratory Medicine, Gävle Hospital, Gävle, Sweden
| | - Karin Jirström
- Division of Oncology and Therapeutic Pathology, Department of Clinical Sciences Lund, Lund, Sweden
| | - Klas Kärre
- Department of Microbiology, Cell and Tumor Biology, Karolinska Institutet, Stockholm, Sweden
| | - Karin Leandersson
- Cancer Immunology, Department of Translational Medicine, Lund University, Skånes University Hospital, Malmö, Sweden
| | - Artur Mezheyeuski
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fredrik Pontén
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Carina Strell
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
Herrington CS, Poulsom R, Koeppen H, Coates PJ. Recent Advances in Pathology: the 2021 Annual Review Issue of The Journal of Pathology. J Pathol 2021; 254:303-306. [PMID: 34097314 DOI: 10.1002/path.5687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 11/10/2022]
Abstract
The 2021 Annual Review Issue of The Journal of Pathology contains 14 invited reviews on current research areas of particular importance in pathology. The subjects included here reflect the broad range of interests covered by the journal, including both basic and applied research fields but always with the aim of improving our understanding of human disease. This year, our reviews encompass the huge impact of the COVID-19 pandemic, the development and application of biomarkers for immune checkpoint inhibitors, recent advances in multiplexing antigen/nucleic acid detection in situ, the use of genomics to aid drug discovery, organoid methodologies in research, the microbiome in cancer, the role of macrophage-stroma interactions in fibrosis, and TGF-β as a driver of fibrosis in multiple pathologies. Other reviews revisit the p53 field and its lack of clinical impact to date, dissect the genetics of mitochondrial diseases, summarise the cells of origin and genetics of sarcomagenesis, provide new data on the role of TRIM28 in tumour predisposition, review our current understanding of cancer stem cell niches, and the function and regulation of p63. The reviews are authored by experts in their field from academia and industry, and provide comprehensive updates of the chosen areas, in which there has been considerable recent progress. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- C Simon Herrington
- Edinburgh Cancer Research Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Richard Poulsom
- The Pathological Society of Great Britain and Ireland, London, UK
| | | | - Philip J Coates
- RECAMO, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| |
Collapse
|