1
|
Elizagaray ML, Barrachina F, Avenatti MC, Bastepe I, Chen A, Odriozola A, Ukairo O, Ros VD, Ottino K, Subiran N, Battistone MA. Chronic inflammation drives epididymal tertiary lymphoid structure formation and autoimmune fertility disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623224. [PMID: 39605691 PMCID: PMC11601424 DOI: 10.1101/2024.11.12.623224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The incomplete understanding of epididymal mucosal immunity is a significant contributing factor to the classification of many male infertility cases as idiopathic. Conditions that disrupt the immune balance in the male reproductive tract, such as vasectomy and infections, can expose sperm to the immune system, leading to increased production of anti-sperm antibodies (ASAs) and subsequent reproductive challenges. Regulatory T cells (Tregs) regulate inflammation and maintain sperm tolerance. In a murine model, we demonstrated that disrupting sperm immunotolerance induces chronic autoimmune responses characterized by antibody production targeting sperm and reproductive tissue autoantigens and unique tissue-specific immune cell signatures in the epididymis and testis. Such inflammatory features impair sperm function, contribute to epididymal damage, and drive sustained male subfertility. Tertiary lymphoid structures (TLSs) were formed within the epididymis after Treg depletion, defined by clusters of heterogenous B and T cells, fibroblasts, and endothelial cells. These ectopic structures perpetuate inflammation and lower the activation threshold for future immune threats. Similar isotypes of autoantibodies were detected in the seminal plasma of infertile patients, suggesting shared mechanistic pathways between mice and humans. Overall, we provide an in-depth understanding of the diverse B- and T-cell dynamics and TLS formation during epididymitis to develop precision-targeted therapies for infertility and chronic inflammation. Additionally, this immunological characterization of the epididymal microenvironment has the potential to identify novel targets for the development of male contraceptives. One Sentence Summary Understanding the epididymal immune cell landscape dynamics aids in developing targeted therapies for infertility and contraception.
Collapse
|
2
|
Chen G, Wang W, Wei X, Chen Y, Peng L, Qu R, Luo Y, He S, Liu Y, Du J, Lu R, Li S, Fan C, Chen S, Dai Y, Yang L. Single-cell transcriptomic analysis reveals that the APP-CD74 axis promotes immunosuppression and progression of testicular tumors. J Pathol 2024; 264:250-269. [PMID: 39161125 DOI: 10.1002/path.6343] [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: 01/24/2024] [Revised: 06/09/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024]
Abstract
Testicular tumors represent the most common malignancy among young men. Nevertheless, the pathogenesis and molecular underpinning of testicular tumors remain largely elusive. We aimed to delineate the intricate intra-tumoral heterogeneity and the network of intercellular communication within the tumor microenvironment. A total of 40,760 single-cell transcriptomes were analyzed, encompassing samples from six individuals with seminomas, two patients with mixed germ cell tumors, one patient with a Leydig cell tumor, and three healthy donors. Five distinct malignant subclusters were identified in the constructed landscape. Among them, malignant 1 and 3 subclusters were associated with a more immunosuppressive state and displayed worse disease-free survival. Further analysis identified that APP-CD74 interactions were significantly strengthened between malignant 1 and 3 subclusters and 14 types of immune subpopulations. In addition, we established an aberrant spermatogenesis trajectory and delineated the global gene alterations of somatic cells in seminoma testes. Sertoli cells were identified as the somatic cell type that differed the most from healthy donors to seminoma testes. Cellular communication between spermatogonial stem cells and Sertoli cells is disturbed in seminoma testes. Our study delineates the intra-tumoral heterogeneity and the tumor immune microenvironment in testicular tumors, offering novel insights for targeted therapy. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Guo Chen
- Department of Urology/Pelvic Floor and Andrology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, PR China
| | - Wei Wang
- Laboratory of Reconstructive Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, PR China
| | - Xin Wei
- Laboratory of Reconstructive Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yulin Chen
- Laboratory of Stem Cell Biology, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, PR China
| | - Liao Peng
- Laboratory of Reconstructive Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, PR China
| | - Rui Qu
- Department of Urology/Pelvic Floor and Andrology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, PR China
| | - Yi Luo
- Laboratory of Reconstructive Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, PR China
| | - Shengyin He
- Department of Urology/Pelvic Floor and Andrology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, PR China
| | - Yugao Liu
- Department of Urology/Pelvic Floor and Andrology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, PR China
| | - Jie Du
- Department of Urology/Pelvic Floor and Andrology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, PR China
| | - Ran Lu
- Laboratory of Stem Cell Biology, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, PR China
| | - Siying Li
- Laboratory of Stem Cell Biology, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, PR China
| | - Chuangwen Fan
- Laboratory of Stem Cell Biology, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, PR China
- Department of Gastrointestinal, Bariatric and Metabolic Surgery, Research Center for Nutrition, Metabolism & Food Safety, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, PR China
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Sujun Chen
- Department of Urology/Pelvic Floor and Andrology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, PR China
| | - Yi Dai
- Department of Urology/Pelvic Floor and Andrology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, PR China
| | - Luo Yang
- Department of Urology/Pelvic Floor and Andrology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
3
|
Castenmiller SM, Kanagasabesan N, Guislain A, Nicolet BP, van Loenen MM, Monkhorst K, Veenhof AA, Smit EF, Hartemink KJ, Haanen JB, de Groot R, Wolkers MC. Tertiary lymphoid structure-related immune infiltrates in NSCLC tumor lesions correlate with low tumor-reactivity of TIL products. Oncoimmunology 2024; 13:2392898. [PMID: 39188755 PMCID: PMC11346574 DOI: 10.1080/2162402x.2024.2392898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/23/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024] Open
Abstract
Adoptive transfer of tumor infiltrating lymphocytes (TIL therapy) has proven highly effective for treating solid cancers, including non-small cell lung cancer (NSCLC). However, not all patients benefit from this therapy for yet unknown reasons. Defining markers that correlate with high tumor-reactivity of the autologous TIL products is thus key for achieving better tailored immunotherapies. We questioned whether the composition of immune cell infiltrates correlated with the tumor-reactivity of expanded TIL products. Unbiased flow cytometry analysis of immune cell infiltrates of 26 early-stage and 20 late-stage NSCLC tumor lesions was used for correlations with the T cell differentiation and activation status, and with the expansion rate and anti-tumor response of generated TIL products. The composition of tumor immune infiltrates was highly variable between patients. Spearman's Rank Correlation revealed that high B cell infiltration negatively correlated with the tumor-reactivity of the patient's expanded TIL products, as defined by cytokine production upon exposure to autologous tumor digest. In-depth analysis revealed that tumor lesions with high B cell infiltrates contained tertiary lymphoid structure (TLS)-related immune infiltrates, including BCL6+ antibody-secreting B cells, IgD+BCL6+ B cells and CXCR5+BLC6+ CD4+ T cells, and higher percentages of naïve CD8+ T cells. In conclusion, the composition of immune cell infiltrates in NSCLC tumors associates with the functionality of the expanded TIL product. Our findings may thus help improve patient selection for TIL therapy.
Collapse
Affiliation(s)
- Suzanne M. Castenmiller
- Sanquin Blood Supply, Division Research Immunotherapy, and Landsteiner Laboratory and Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Nandhini Kanagasabesan
- Sanquin Blood Supply, Division Research Immunotherapy, and Landsteiner Laboratory and Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Aurélie Guislain
- Sanquin Blood Supply, Division Research Immunotherapy, and Landsteiner Laboratory and Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Benoît P. Nicolet
- Sanquin Blood Supply, Division Research Immunotherapy, and Landsteiner Laboratory and Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Marleen M. van Loenen
- Sanquin Blood Supply, Division Research Immunotherapy, and Landsteiner Laboratory and Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Kim Monkhorst
- Department of Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AvL), Amsterdam, The Netherlands
| | - Alexander A.F.A. Veenhof
- Department of Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AvL), Amsterdam, The Netherlands
| | - Egbert F. Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AvL), Amsterdam, The Netherlands
- Department of Pulmonology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Koen J. Hartemink
- Department of Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AvL), Amsterdam, The Netherlands
| | - John B.A.G. Haanen
- Division of Medical Oncology and Division of Molecular Oncology and Immunology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AvL), Amsterdam, The Netherlands
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Head of Melanoma Clinic, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Rosa de Groot
- Sanquin Blood Supply, Division Research Immunotherapy, and Landsteiner Laboratory and Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
- Department of Hematology, LUMC, Leiden, The Netherlands
| | - Monika C. Wolkers
- Sanquin Blood Supply, Division Research Immunotherapy, and Landsteiner Laboratory and Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam, Netherlands
- Oncode Institute, Utrecht, The Netherlands
| |
Collapse
|
4
|
Cho YJ, Hwang I, Park S, Lee S, Kang SY, Kim MJ, Ahn S, Kim KM. Prognostic Effect of Tertiary Lymphoid Structures in Epstein-Barr Virus-Associated Gastric Carcinomas Measured by Digital Image Analysis. J Transl Med 2024; 104:102071. [PMID: 38677591 DOI: 10.1016/j.labinv.2024.102071] [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: 10/03/2023] [Revised: 02/25/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024] Open
Abstract
Epstein-Barr virus-associated gastric carcinoma (EBVaGC) is characterized by prominent tumor-infiltrating lymphocytes (TILs) and has a favorable prognosis. Tertiary lymphoid structures (TLS), characterized by ectopic aggregated lymphocytes with high-endothelial venules (HEV), are associated with favorable outcomes in various solid tumors. We hypothesized that EBVaGC, characterized by intense TILs, may be closely associated with TLS or HEV. To test this hypothesis, we digitally analyzed the TLS, HEV, and TILs in 73 surgically resected advanced EBVaGCs. For HEV, dual MECA-79 and CD31 dual immunohistochemistry were performed, and the ectopic expression of MECA-79 in tumor cells was measured. In 73 patients with EBVaGC, a high-TLS ratio was found in 29 (39.7%) cases, high-tumor-associated HEV density in 44 (60.3%) cases, and high-CD8+ TIL density in 38 (52.1%) cases. Ectopic tumor expression of MECA-79 was observed in 36 patients (49.3%) cases. A low-TLS ratio and tumor-associated HEV density were significantly associated with lymph node metastasis (P = .005 and .042, respectively). Ectopic MECA-79 expression was significantly associated with lymph node metastasis (P = .003). Patients with a low-TLS ratio (P = .038), low-HEV density (P = .042), and ectopic tumor MECA-79 expression (P = .032) had significantly worse prognoses. In conclusion, TLS ratio and HEV density affect the survival of patients with EBVaGC and may be related to the immune response that interrupts lymph node metastasis.
Collapse
Affiliation(s)
- Yun Joo Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Inwoo Hwang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Suho Park
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Somin Lee
- Center for Companion Diagnostics, Precision Medicine Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - So Young Kang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Min-Ji Kim
- Biomedical Statistics Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Soomin Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Center for Companion Diagnostics, Precision Medicine Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
5
|
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
|
6
|
Groen-van Schooten TS, Franco Fernandez R, van Grieken NCT, Bos EN, Seidel J, Saris J, Martínez-Ciarpaglini C, Fleitas TC, Thommen DS, de Gruijl TD, Grootjans J, Derks S. Mapping the complexity and diversity of tertiary lymphoid structures in primary and peritoneal metastatic gastric cancer. J Immunother Cancer 2024; 12:e009243. [PMID: 38955417 PMCID: PMC11218001 DOI: 10.1136/jitc-2024-009243] [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: 06/13/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Tertiary lymphoid structures (TLSs) are thought to stimulate antitumor immunity and positively impact prognosis and response to immune checkpoint blockade. In gastric cancers (GCs), however, TLSs are predominantly found in GC with poor prognosis and limited treatment response. We, therefore, hypothesize that immune cell composition and function of TLS depends on tumor location and the tumor immune environment. METHODS Spatial transcriptomics and immunohistochemistry were used to characterize the phenotype of CD45+ immune cells inside and outside of TLS using archival resection specimens from GC primary tumors and peritoneal metastases. RESULTS We identified significant intrapatient and interpatient diversity of the cellular composition and maturation status of TLS in GC. Tumor location (primary vs metastatic site) accounted for the majority of differences in TLS maturity, as TLS in peritoneal metastases were predominantly immature. This was associated with higher levels of tumor-infiltrating macrophages and Tregs and less plasma cells compared with tumors with mature TLS. Furthermore, mature TLSs were characterized by overexpression of antitumor immune pathways such as B cell-related pathways, MHC class II antigen presentation while immature TLS were associated with protumor pathways, including T cell exhaustion and enhancement of DNA repair pathways in the corresponding cancer. CONCLUSION The observation that GC-derived peritoneal metastases often contain immature TLS which are associated with immune suppressive regulatory tumor-infiltrating leucocytes, is in keeping with the lack of response to immune checkpoint blockade and the poor prognostic features of peritoneal metastatic GC, which needs to be taken into account when optimizing immunomodulatory strategies for metastatic GC.
Collapse
Affiliation(s)
- Tessa S Groen-van Schooten
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Rosalia Franco Fernandez
- Oncode Institute, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology & Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Nicole C T van Grieken
- Cancer Biology and Immunology, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
| | - Emma N Bos
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Jens Seidel
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Job Saris
- Oncode Institute, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology & Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | | | | | - Daniela S Thommen
- Oncode Institute, Amsterdam, The Netherlands
- Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Joep Grootjans
- Oncode Institute, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology & Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Sarah Derks
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Wang S, Wang H, Li C, Liu B, He S, Tu C. Tertiary lymphoid structures in cancer: immune mechanisms and clinical implications. MedComm (Beijing) 2024; 5:e489. [PMID: 38469550 PMCID: PMC10925885 DOI: 10.1002/mco2.489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 03/13/2024] Open
Abstract
Cancer is a major cause of death globally, and traditional treatments often have limited efficacy and adverse effects. Immunotherapy has shown promise in various malignancies but is less effective in tumors with low immunogenicity or immunosuppressive microenvironment, especially sarcomas. Tertiary lymphoid structures (TLSs) have been associated with a favorable response to immunotherapy and improved survival in cancer patients. However, the immunological mechanisms and clinical significance of TLS in malignant tumors are not fully understood. In this review, we elucidate the composition, neogenesis, and immune characteristics of TLS in tumors, as well as the inflammatory response in cancer development. An in-depth discussion of the unique immune characteristics of TLSs in lung cancer, breast cancer, melanoma, and soft tissue sarcomas will be presented. Additionally, the therapeutic implications of TLS, including its role as a marker of therapeutic response and prognosis, and strategies to promote TLS formation and maturation will be explored. Overall, we aim to provide a comprehensive understanding of the role of TLS in the tumor immune microenvironment and suggest potential interventions for cancer treatment.
Collapse
Affiliation(s)
- Siyu Wang
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Xiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Hua Wang
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Chenbei Li
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Binfeng Liu
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Shasha He
- Department of OncologyThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Chao Tu
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Shenzhen Research Institute of Central South UniversityGuangdongChina
- Changsha Medical UniversityChangshaChina
| |
Collapse
|
8
|
Chen H, Li G, Cui Y, Zhang Q, Li B, Liu X. High endothelial venules in intracranial germinomas: Implications for lymphocytes infiltration. Cancer Med 2023; 12:5450-5460. [PMID: 36259639 PMCID: PMC10028053 DOI: 10.1002/cam4.5367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/27/2022] [Accepted: 10/07/2022] [Indexed: 11/12/2022] Open
Abstract
PURPOSE Reactive lymphocytes are substantial components of germinoma, which are believed to be related to the favorable prognosis of this intracranial tumor and better response to immunotherapy. However, the mechanisms managing the recruitment of lymphocytes are poorly understood. High endothelial venules (HEVs) are specialized blood vessels that play key roles in lymphocyte trafficking in Lymph nodes. These vessels are associated with lymphocyte infiltration in chronic inflammatory diseases and various malignant tumors, but their distribution and implications in germinoma are unknown. This study aimed to investigate the distribution and implications of HEVs in intracranial germinomas. METHODS We investigated the presence and distribution of HEVs in 42 germinomas by immunohistochemical staining of peripheral node addressin (PNAd) and transmission electron microscopic examination. The correlation of the densities of HEVs with the extent of T and B lymphocyte infiltration and several clinicopathological characteristics were also analyzed to determine whether HEVs are responsible for lymphocyte recruitment and their roles in anti-tumor immunity in germinoma. RESULTS PNAd-positive HEVs were detected in 31% (13/42) of germinomas, and their presence correlated with abundant infiltrating CD3+ T cells, CD20 + B cells and CD8+ cytotoxic T lymphocytes (p = 0.0410, 0.0023, and 0.0061, respectively). Higher HEVs density was also correlated with several clinicopathological parameters, which are recognized indicators for favorable prognosis in germinomas, including typical tumor location (p = 0.0093), lower tumor cell content (p = 0.0428), and younger age at diagnosis (p = 0.0121). Furthermore, bioinformatics analysis showed HEVs-associated genes mainly enriched in immune-related Gene Ontology terms, including innate immune response, inflammatory response, and B cell receptor signaling pathway. The xCell analysis revealed that germinomas with higher HEVs enrichment scores had increased levels of the immune score, microenvironment score, dendritic cells, CD8+ central memory T-cells, CD4+ memory T-cells, and B-cells. CONCLUSIONS Our findings indicate that HEVs could contribute to lymphocyte recruitment in germinomas, thus may serve as a predictor of favorable prognosis and better response to immunotherapy in this intracranial tumor.
Collapse
Affiliation(s)
- Huiyuan Chen
- Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, People's Republic of China
| | - Guilin Li
- Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, People's Republic of China
| | - Yun Cui
- Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, People's Republic of China
| | - Qi Zhang
- Department of Ultrastructure Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, People's Republic of China
| | - Bo Li
- Department of Radiation Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xing Liu
- Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
9
|
Zhang Q, Wu S. Tertiary lymphoid structures are critical for cancer prognosis and therapeutic response. Front Immunol 2023; 13:1063711. [PMID: 36713409 PMCID: PMC9875059 DOI: 10.3389/fimmu.2022.1063711] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
Tertiary lymphoid structures (TLSs) are ectopic lymphocyte aggregates that form at sites of chronic inflammation, including cancers, in non-lymphoid tissues. Although the formation of TLSs is similar to that of secondary lymphoid organs, the pathogenic factors leading to TLS formation in cancerous tissues and the mechanisms underlying the role of these structures in the intra-tumoral adaptive antitumor immune response are not fully understood. The presence of TLSs may impact patient prognosis and treatment outcomes. This review examines the current understanding of TLSs in cancers, including their composition and formation as well as their potential to predict prognosis and therapeutic efficacy. We also summarize strategies to induce TLS formation for cancer treatment.
Collapse
Affiliation(s)
| | - Suhui Wu
- Department of Obstetrics and Gynecology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
10
|
Rodriguez AB, Parriott G, Engelhard VH. Tumor necrosis factor receptor regulation of peripheral node addressin biosynthetic components in tumor endothelial cells. Front Immunol 2022; 13:1009306. [PMID: 36189308 PMCID: PMC9520236 DOI: 10.3389/fimmu.2022.1009306] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022] Open
Abstract
Tumor-associated tertiary lymphoid structures are ectopic lymphoid aggregates that have considerable morphological, cellular, and molecular similarity to secondary lymphoid organs, particularly lymph nodes. Tumor vessels expressing peripheral node addressin (PNAd) are hallmark features of these structures. Previous work from our laboratory demonstrated that PNAd is displayed on intratumoral vasculature of murine tumors, and its expression is controlled by the engagement of lymphotoxin-α3, secreted by effector CD8 T cells, with tumor necrosis factor receptors (TNFR) on tumor endothelial cells (TEC). The goals of the present work were: 1) to identify differences in expression of genes encoding the scaffolding proteins and glycosyl transferases associated with PNAd biosynthesis in TEC and lymph node blood endothelial cells (LN BEC); and 2) to determine which of these PNAd associated components are regulated by TNFR signaling. We found that the same genes encoding scaffolding proteins and glycosyl transferases were upregulated in PNAd+ LN BEC and PNAd+ TEC relative to their PNAdneg counterparts. The lower level of PNAd expression on TEC vs LN BEC was associated with relatively lower expression of these genes, particularly the carbohydrate sulfotransferase Chst4. Loss of PNAd on TEC in the absence of TNFR signaling was associated with lack of upregulation of these same genes. A small subset of PNAd+ TEC remaining in the absence of TNFR signaling showed normal upregulation of a subset of these genes, but reduced upregulation of genes encoding the scaffolding proteins podocalyxin and nepmucin, and carbohydrate sulfotransferase Chst2. Lastly, we found that checkpoint immunotherapy augmented both the fraction of TEC expressing PNAd and their surface level of this ligand. This work points to strong similarities in the regulation of PNAd expression on TEC by TNFR signaling and on LN BEC by lymphotoxin-β receptor signaling, and provides a platform for the development of novel strategies that manipulate PNAd expression on tumor vasculature as an element of cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Victor H. Engelhard
- Carter Immunology Center and Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
- *Correspondence: Victor H. Engelhard,
| |
Collapse
|
11
|
Murahashi M, Kogami A, Muramoto A, Hoshino H, Akama TO, Mitoma J, Goi T, Hirayama A, Okamura T, Nagaya T, Kobayashi M. Vascular E-selectin Expression Detected in Formalin-fixed, Paraffin-embedded Sections With an E-selectin Monoclonal Antibody Correlates With Ulcerative Colitis Activity. J Histochem Cytochem 2022; 70:299-310. [PMID: 35253509 PMCID: PMC8971687 DOI: 10.1369/00221554221085336] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
It is widely accepted that E-selectin, an inducible endothelial cell adhesion molecule, plays a critical role in the initial step of neutrophil recruitment to sites of acute inflammation. However, immunohistological analysis of E-selectin has been hampered by lack of E-selectin-specific monoclonal antibodies that can stain formalin-fixed, paraffin-embedded (FFPE) tissue sections. Here, we employed E-selectin•IgM (a soluble form of E-selectin) as immunogen, and then, after negative selection with L-selectin•IgM and P-selectin•IgM and screening of FFPE sections of both COS-1 cells overexpressing E-selectin and acute appendicitis tissues, we successfully generated an E-selectin-specific monoclonal antibody capable of staining FFPE tissue sections. We used this antibody, designated U12-12, to perform quantitative immunohistological analysis of 390 colonic mucosal biopsy specimens representing ulcerative colitis. We found that the higher the histological disease activity, the greater the number of vessels expressing E-selectin, an observation consistent with previous analyses of frozen tissue sections. Furthermore, in active ulcerative colitis, E-selectin-expressing vessels contained neutrophils attached to endothelial cells, presumably in the process of extravasation, which eventually could cause epithelial damage. These results overall indicate that U12-12 is effective for E-selectin immunohistochemistry in archived FFPE samples representing various human diseases.
Collapse
Affiliation(s)
| | | | | | | | - Tomoya O Akama
- Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan; Department of Pharmacology, Kansai Medical University, Hirakata, Japan
| | - Junya Mitoma
- Department of Medical Life Sciences, School of Medical Life Sciences, Kyushu University of Health and Welfare, Nobeoka, Japan
| | | | - Atsuhiro Hirayama
- Division of Gastroenterology and Hepatology, Department of Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takuma Okamura
- Division of Gastroenterology and Hepatology, Department of Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Tadanobu Nagaya
- Division of Gastroenterology and Hepatology, Department of Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | | |
Collapse
|
12
|
Kogami A, Fukushima M, Hoshino H, Komeno T, Okoshi T, Murahashi M, Akama TO, Mitoma J, Ohtani H, Kobayashi M. The Conspicuousness of High Endothelial Venules in Angioimmunoblastic T-cell Lymphoma Is Due to Increased Cross-sectional Area, Not Increased Distribution Density. J Histochem Cytochem 2021; 69:645-657. [PMID: 34617807 DOI: 10.1369/00221554211048551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is a T-cell lymphoma of follicular helper T-cell origin. Histologically, neoplastic T-cells proliferate to form clusters adjacent to or between arborizing high endothelial venules (HEVs). HEVs in normal lymph nodes express sulfated glycans called peripheral lymph node addressin (PNAd); however, it remains unclear whether PNAd is also expressed on HEVs in AITL. Furthermore, although it is widely accepted that HEVs are conspicuous in AITL due to their proliferation, quantitative histological support for this concept is lacking. To investigate these issues, we employed monoclonal antibodies recognizing PNAd, namely, MECA-79, HECA-452, and 297-11A, and performed quantitative immunohistochemical analysis of HEVs in 36 AITL-affected and 67 normal lymph nodes. Staining with all three antibodies confirmed that AITL HEVs express PNAd. Moreover, AITL HEVs were bound calcium-dependently by L-selectin-IgM fusion proteins, indicating that they function in the recruitment of L-selectin-expressing lymphocytes. Unexpectedly, HEV distribution density was not increased but rather decreased in AITL compared with normal lymph nodes, but HEV cross-sectional area in AITL was significantly greater than that seen in normal lymph nodes. Overall, these results indicate that the prominence of AITL HEVs is likely due to increased cross-sectional area rather than increased distribution density.
Collapse
Affiliation(s)
- Akiya Kogami
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Mana Fukushima
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan.,Omachi Municipal General Hospital, Omachi, Japan
| | - Hitomi Hoshino
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Takuya Komeno
- Department of Hematology, National Hospital Organization Mito Medical Center, Ibaraki, Japan
| | - Tadakazu Okoshi
- Department of Pathology, Japanese Red Cross Fukui Hospital, Fukui, Japan
| | - Masataka Murahashi
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Tomoya O Akama
- Department of Pharmacology, Kansai Medical University, Hirakata, Japan
| | - Junya Mitoma
- Department of Medical Life Sciences, School of Medical Life Sciences, Kyushu University of Health and Welfare, Nobeoka, Japan
| | - Haruo Ohtani
- Department of Pathology, Mito Saiseikai General Hospital, Mito, Japan
| | - Motohiro Kobayashi
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan.,Omachi Municipal General Hospital, Omachi, Japan
| |
Collapse
|
13
|
Gondo N, Sakai Y, Zhang Z, Hato Y, Kuzushima K, Phimsen S, Kawashima Y, Kuroda M, Suzuki M, Okada S, Iwata H, Toyama T, Rezano A, Kuwahara K. Increased chemosensitivity via BRCA2-independent DNA damage in DSS1- and PCID2-depleted breast carcinomas. J Transl Med 2021; 101:1048-1059. [PMID: 34031538 DOI: 10.1038/s41374-021-00613-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/08/2021] [Accepted: 05/09/2021] [Indexed: 11/09/2022] Open
Abstract
Breast cancer, the most common malignancy among women, is closely associated with mutations in the tumor suppressor gene BRCA. DSS1, a component of the TRanscription-EXport-2 (TREX-2) complex involved in transcription and mRNA nuclear export, stabilizes BRCA2 expression. DSS1 is also related to poor prognosis in patients with breast cancer owing to the induction of chemoresistance. Recently, BRCA2 was shown to be associated with the TREX-2 component PCID2, which prevents DNA:RNA hybrid R-loop formation and transcription-coupled DNA damage. This study aimed to elucidate the involvement of these TREX-2 components and BRCA2 in the chemosensitivity of breast carcinomas. Our results showed that compared with that in normal breast tissues, DSS1 expression was upregulated in human breast carcinoma, whereas PCID2 expression was comparable between normal and malignant tissues. We then compared patient survival time among groups divided by high or low expressions of DSS1, BRCA2, and PCID2. Increased DSS1 expression was significantly correlated with poor prognosis in recurrence-free survival time, whereas no differences were detected in the high and low BRCA2 and PCID2 expression groups. We performed in vitro analyses, including propidium iodide nuclear staining, single-cell gel electrophoresis, and clonogenic survival assays, using breast carcinoma cell lines. The results confirmed that DSS1 depletion significantly increased chemosensitivity, whereas overexpression conferred chemoresistance to breast cancer cell lines; however, BRCA2 expression did not affect chemosensitivity. Similar to DSS1, PCID2 expression was also inversely correlated with chemosensitivity. These results strongly suggest that DSS1 and PCID2 depletion is closely associated with increased chemosensitivity via BRCA2-independent DNA damage. Together with the finding that DSS1 is not highly expressed in normal breast tissues, these results demonstrate that DSS1 depletion confers a druggable trait and may contribute to the development of novel chemotherapeutic strategies to treat DSS1-depleted breast carcinomas independent of BRCA2 mutations.
Collapse
Affiliation(s)
- Naomi Gondo
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cellular Oncology, Department of Cancer Genetics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yasuhiro Sakai
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Zhenhuan Zhang
- Radiation Oncology Department, University of Florida, Gainesville, FL, USA
| | - Yukari Hato
- Department of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kiyotaka Kuzushima
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cellular Oncology, Department of Cancer Genetics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Suchada Phimsen
- Faculty of Medical Science, Department of Biochemistry, Naresuan University, Phitsanulok, Thailand
| | - Yoshiaki Kawashima
- Department of Pathology, Fujita Health University Hospital, Toyoake, Japan
| | - Makoto Kuroda
- Department of Pathology, Fujita Health University Okazaki Medical Center, Okazaki, Japan
| | - Motoshi Suzuki
- Department of Molecular Oncology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Retroviral Infection, Kumamoto University, Kumamoto, Japan
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Tatsuya Toyama
- Department of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Andri Rezano
- Division of Cell Biology, Faculty of Medicine, Department of Biomedical Sciences, Universitas Padjadjaran, West Java, Indonesia.
| | - Kazuhiko Kuwahara
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya, Japan.
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Toyoake, Japan.
| |
Collapse
|
14
|
Recent Advances in Glioma Therapy: Combining Vascular Normalization and Immune Checkpoint Blockade. Cancers (Basel) 2021; 13:cancers13153686. [PMID: 34359588 PMCID: PMC8345045 DOI: 10.3390/cancers13153686] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma (GBM) accounts for more than 50% of all primary malignancies of the brain. Current standard treatment regimen for GBM includes maximal surgical resection followed by radiation and adjuvant chemotherapy. However, due to the heterogeneity of the tumor cells, tumor recurrence is often inevitable. The prognosis of patients with glioma is, thus, dismal. Glioma is a highly angiogenic tumor yet immunologically cold. As such, evolving studies have focused on designing strategies that specifically target the tyrosine kinase receptors of angiokines and encourage immune infiltration. Recent promising results from immunotherapies on other cancer types have prompted further investigations of this therapy in GBM. In this article, we reviewed the pathological angiogenesis and immune reactivity in glioma, as well as its target for drug development, and we discussed future directions in glioma therapy.
Collapse
|
15
|
Blanchard L, Girard JP. High endothelial venules (HEVs) in immunity, inflammation and cancer. Angiogenesis 2021; 24:719-753. [PMID: 33956259 PMCID: PMC8487881 DOI: 10.1007/s10456-021-09792-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022]
Abstract
High endothelial venules (HEVs) are specialized blood vessels mediating lymphocyte trafficking to lymph nodes (LNs) and other secondary lymphoid organs. By supporting high levels of lymphocyte extravasation from the blood, HEVs play an essential role in lymphocyte recirculation and immune surveillance for foreign invaders (bacterial and viral infections) and alterations in the body’s own cells (neoantigens in cancer). The HEV network expands during inflammation in immune-stimulated LNs and is profoundly remodeled in metastatic and tumor-draining LNs. HEV-like blood vessels expressing high levels of the HEV-specific sulfated MECA-79 antigens are induced in non-lymphoid tissues at sites of chronic inflammation in many human inflammatory and allergic diseases, including rheumatoid arthritis, Crohn’s disease, allergic rhinitis and asthma. Such vessels are believed to contribute to the amplification and maintenance of chronic inflammation. MECA-79+ tumor-associated HEVs (TA-HEVs) are frequently found in human tumors in CD3+ T cell-rich areas or CD20+ B-cell rich tertiary lymphoid structures (TLSs). TA-HEVs have been proposed to play important roles in lymphocyte entry into tumors, a process essential for successful antitumor immunity and lymphocyte-mediated cancer immunotherapy with immune checkpoint inhibitors, vaccines or adoptive T cell therapy. In this review, we highlight the phenotype and function of HEVs in homeostatic, inflamed and tumor-draining lymph nodes, and those of HEV-like blood vessels in chronic inflammatory diseases. Furthermore, we discuss the role and regulation of TA-HEVs in human cancer and mouse tumor models.
Collapse
Affiliation(s)
- Lucas Blanchard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
16
|
High Endothelial Venule with Concomitant High CD8+ Tumor-Infiltrating Lymphocytes Is Associated with a Favorable Prognosis in Resected Gastric Cancer. J Clin Med 2020; 9:jcm9082628. [PMID: 32823631 PMCID: PMC7464373 DOI: 10.3390/jcm9082628] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/24/2022] Open
Abstract
CD8+ tumor-infiltrating lymphocytes (TILs) play a major role in antitumor immunity. High endothelial venules (HEVs) are related to diverse immune cells in solid tumors. We analyzed CD8+ and Foxp3+ TILs in combination with HEVs to determine their prognostic role in advanced gastric cancer (AGC). We enrolled 157 patients with AGC in this study. The densities of CD8+ TILs and Foxp3+ TILs were calculated using immunohistochemical staining. HEVs were evaluated by MECA-79 expression. HEVs were identified in 60 (38.2%) cases and was significantly associated with an increased number of CD8+ TILs (p = 0.027) but not of Foxp3+ TILs (p = 0.455) and CD20+ TILs (p = 0.163). A high CD8+/HEV+ level was significantly associated with nodal metastasis (p = 0.048). In survival analysis, patients with high CD8+/HEV+ levels demonstrated the longest overall survival (OS) (p = 0.015). Furthermore, a high CD8+/HEV+ level was an independent prognostic factor in AGC (p = 0.011; hazard ratio (HR) = 0.435; 95% confidence interval (CI) = 0.245–0.837). HEVs were found to play an important role in antitumor immunity associated with CD8+ TILs in AGC. This analysis of HEVs and CD8+ TILs helps stratify patients with AGC and sheds light on tumor immunity.
Collapse
|
17
|
Sakai Y, Rezano A, Okada S, Ohtsuki T, Kawashima Y, Tsukamoto T, Suzuki M, Kohara M, Takeya M, Sakaguchi N, Kuwahara K. A Novel Cytological Model of B-Cell/Macrophage Biphenotypic Cell Hodgkin Lymphoma in Ganp-Transgenic Mice. Cancers (Basel) 2020; 12:cancers12010204. [PMID: 31947626 PMCID: PMC7017268 DOI: 10.3390/cancers12010204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/09/2020] [Accepted: 01/11/2020] [Indexed: 11/20/2022] Open
Abstract
Hodgkin lymphoma (HL) is one of the most difficult neoplasms in terms of cytopathological research owing to the lack of established cytological murine models. Although HL is believed to be of lymphoid germinal center B-cell origin, HL cells exhibit unique biphenotypic characteristics of B cells and macrophages. B-cell/macrophage biphenotypic cells have also been identified in the spleen of Lyn-deficient mice. Moreover, Lyn-targeting germinal center-associated nuclear protein (GANP)-transgenic mice (Ig-ganpTg mice) spontaneously develop a lymphoid tumor. We aimed to investigate whether the lymphoid tumor developed in Ig-ganpTg mice exhibit biphenotypic characteristics of B cells/macrophages that correspond to human HL. Here, we demonstrated GANP overexpression in human HL cells and found that it may regulate transdifferentiation between B cells and macrophages. We also demonstrated that tumors were comparable with B-cell/macrophage biphenotypic Hodgkinoid lymphomas. The tumor cells expressed macrophage-related F4/80, CD68, and CD204 as well as cytoplasmic B220 and µ-/κ-chains; in addition, these cells exhibited phagocytic activity. These cells also expressed transcripts of CD30; c-fms; and the cytokines monocyte chemoattractant protein (MCP)-1, MCP-5, RANTES, tumor necrosis factor-α and thrombopoietin associated with macrophages as well as granulocyte/macrophage colony-stimulating factor, interleukin (IL)-4, IL-10, IL-12, and IL-13. Ig-ganpTg mice represent a novel cytological model for the study of cytopathological etiology and oncogenesis of HL.
Collapse
Affiliation(s)
- Yasuhiro Sakai
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Aichi 470-1192, Japan; (Y.S.); (Y.K.); (T.T.)
| | - Andri Rezano
- Department of Biomedical Sciences, Division of Cell Biology, Faculty of Medicine, Universitas Padjadjaran, West Java 45363, Indonesia;
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Retroviral Infection, Kumamoto University, Kumamoto 860-0811, Japan;
| | - Takahiro Ohtsuki
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (T.O.); (M.K.)
| | - Yoshiaki Kawashima
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Aichi 470-1192, Japan; (Y.S.); (Y.K.); (T.T.)
| | - Tetsuya Tsukamoto
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Aichi 470-1192, Japan; (Y.S.); (Y.K.); (T.T.)
| | - Motoshi Suzuki
- Department of Molecular Oncology, Fujita Health University School of Medicine, Aichi 470-1192, Japan;
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (T.O.); (M.K.)
| | - Motohiro Takeya
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan;
| | - Nobuo Sakaguchi
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan;
| | - Kazuhiko Kuwahara
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Aichi 470-1192, Japan; (Y.S.); (Y.K.); (T.T.)
- Correspondence: ; Tel.: +81-562-93-2319; Fax: +81-562-95-3761
| |
Collapse
|
18
|
Jennewein L, Bartsch G, Gust K, Kvasnicka HM, Haferkamp A, Blaheta R, Mittelbronn M, Harter PN, Mani J. Increased tumor vascularization is associated with the amount of immune competent PD-1 positive cells in testicular germ cell tumors. Oncol Lett 2018; 15:9852-9860. [PMID: 29928359 PMCID: PMC6004709 DOI: 10.3892/ol.2018.8597] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/22/2018] [Indexed: 12/12/2022] Open
Abstract
Testicular germ cell cancer in a metastatic state is curable with a cisplatin-based first line chemotherapy. However, 10-15% of these patients are resistant to first line chemotherapy and are thus left with only palliative options. Immunotherapies and inhibition of angiogenesis used in multiple types of cancer; however, the molecular context of angiogenesis and immune checkpoints in the development and progression of testicular cancers is still unknown. Therefore, the present study performed tissue micro array based analysis of 84 patients with immunohistochemistry of programmed cell death protein 1 (PD-1), programmed cell death ligand 1 (PD-L1) and vascular endothelial growth factor receptor 2 (VEGFR2) of testicular cancer and corresponding normal appearing testis tissue, matching the results with clinical data. The results demonstrated that PD-L1 was significantly upregulated in testicular tumors and that PD-1 positive cells significantly infiltrated the testicular tumor when compared with normal testicular tissue. VEGFR2 was significantly upregulated in testicular cancer. It was indicated that PD-1 expressing cytotoxic cells may require pathologic tumor vessels to pass the blood-testis-barrier in order to migrate into the tumor. Notably, when matching the clinical data for PD-1, PD-L1 and VEGFR2 there were no differences in expression in the different International Germ Cell Cancer Collaborative Group stages of non-seminoma. These data suggested that the anti-PD-1/PD-L1 immunotherapy and the anti-angiogenic therapy, sequentially or in combination, may be a promising option in the treatment of testicular cancer.
Collapse
Affiliation(s)
- Lukas Jennewein
- Neurological Institute (Edinger Institute), Goethe University, D-60528 Frankfurt/Main, Germany.,Department of Obstetrics and Gynecology, Goethe University, D-60590 Frankfurt/Main, Germany
| | - Georg Bartsch
- Department of Urology, Johannes Gutenberg University, D-55131 Mainz, Germany
| | - Kilian Gust
- Department of Urology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Hans-Michael Kvasnicka
- Senckenberg Institute of Pathology, Goethe University of Frankfurt/Main, D-60590 Frankfurt/Main, Germany.,German Cancer Consortium (DKTK) and German Consortium for Translational Cancer Research (DKTK), D-69120 Heidelberg, Germany
| | - Axel Haferkamp
- Department of Urology, Johannes Gutenberg University, D-55131 Mainz, Germany
| | - Roman Blaheta
- Department of Urology, Goethe University, D-60590 Frankfurt/Main, Germany
| | | | - Patrick N Harter
- Neurological Institute (Edinger Institute), Goethe University, D-60528 Frankfurt/Main, Germany.,German Cancer Consortium (DKTK) and German Consortium for Translational Cancer Research (DKTK), D-69120 Heidelberg, Germany
| | - Jens Mani
- Department of Urology, Goethe University, D-60590 Frankfurt/Main, Germany
| |
Collapse
|
19
|
Engelhard VH, Rodriguez AB, Mauldin IS, Woods AN, Peske JD, Slingluff CL. Immune Cell Infiltration and Tertiary Lymphoid Structures as Determinants of Antitumor Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:432-442. [PMID: 29311385 PMCID: PMC5777336 DOI: 10.4049/jimmunol.1701269] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/19/2017] [Indexed: 12/22/2022]
Abstract
Limited representation of intratumoral immune cells is a major barrier to tumor control. However, simply enhancing immune responses in tumor-draining lymph nodes or through adoptive transfer may not overcome the limited ability of tumor vasculature to support effector infiltration. An alternative is to promote a sustained immune response intratumorally. This idea has gained traction with the observation that many tumors are associated with tertiary lymphoid structures (TLS), which organizationally resemble lymph nodes. These peri- and intratumoral structures are usually, but not always, associated with positive prognoses in patients. Preclinical and clinical data support a role for TLS in modulating immunity in the tumor microenvironment. However, there appear to be varied functions of TLS, potentially based on their structure or location in relation to the tumor or the origin or location of the tumor itself. Understanding more about TLS development, composition, and function may offer new therapeutic opportunities to modulate antitumor immunity.
Collapse
Affiliation(s)
- Victor H Engelhard
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908;
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - Anthony B Rodriguez
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - Ileana S Mauldin
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Amber N Woods
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - J David Peske
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - Craig L Slingluff
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908
| |
Collapse
|
20
|
Colbeck EJ, Ager A, Gallimore A, Jones GW. Tertiary Lymphoid Structures in Cancer: Drivers of Antitumor Immunity, Immunosuppression, or Bystander Sentinels in Disease? Front Immunol 2017; 8:1830. [PMID: 29312327 PMCID: PMC5742143 DOI: 10.3389/fimmu.2017.01830] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/04/2017] [Indexed: 12/16/2022] Open
Abstract
Secondary lymphoid organs are integral to initiation and execution of adaptive immune responses. These organs provide a setting for interactions between antigen-specific lymphocytes and antigen-presenting cells recruited from local infected or inflamed tissues. Secondary lymphoid organs develop as a part of a genetically preprogrammed process during embryogenesis. However, organogenesis of secondary lymphoid tissues can also be recapitulated in adulthood during de novo lymphoid neogenesis of tertiary lymphoid structures (TLSs). These ectopic lymphoid-like structures form in the inflamed tissues afflicted by various pathological conditions, including cancer, autoimmunity, infection, or allograft rejection. Studies are beginning to shed light on the function of such structures in different disease settings, raising important questions regarding their contribution to progression or resolution of disease. Data show an association between the tumor-associated TLSs and a favorable prognosis in various types of human cancer, attracting the speculation that TLSs support effective local antitumor immune responses. However, definitive evidence for the role for TLSs in fostering immune responses in vivo are lacking, with current data remaining largely correlative by nature. In fact, some more recent studies have even demonstrated an immunosuppressive, tumor-promoting role for cancer-associated TLSs. In this review, we will discuss what is known about the development of cancer-associated TLSs and the current understanding of their potential role in the antitumor immune response.
Collapse
Affiliation(s)
| | - Ann Ager
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Awen Gallimore
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Gareth Wyn Jones
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
21
|
Low S, Sakai Y, Hoshino H, Hirokawa M, Kawashima H, Higuchi K, Imamura Y, Kobayashi M. High endothelial venule-like vessels and lymphocyte recruitment in diffuse sclerosing variant of papillary thyroid carcinoma. Pathology 2016; 48:666-674. [PMID: 27956273 DOI: 10.1016/j.pathol.2016.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/16/2016] [Accepted: 08/08/2016] [Indexed: 01/19/2023]
Abstract
Diffuse sclerosing variant of papillary thyroid carcinoma (DSPTC) is a rare subtype of papillary thyroid carcinoma with a high incidence of lymph node metastasis. One of its characteristic histological features is the presence of dense lymphocyte infiltrates; however, how these lymphocytes are recruited in this pathological setting remains unclear. Here, we analysed 17 DSPTC cases immunohistologically for cell adhesion molecules expressed on endothelial cells. We found that venules morphologically similar to high endothelial venules (HEVs) in secondary lymphoid organs were induced in lymphoid aggregates in DSPTC, and such HEV-like vessels expressed 6-sulfo sialyl Lewis X (sLeX) glycans as well as intercellular adhesion molecule 1 (ICAM-1). Triple immunohistochemistry revealed that CD8+ cytotoxic T cells were the major lymphocyte subset attached to the luminal surface of HEV-like vessels. sLeX-type glycans were also expressed on DSPTC carcinoma cells, which in binding assays were decorated with E-selectin•IgM chimaeras calcium-dependently. These findings collectively suggest that 6-sulfo sLeX glycans, together with ICAM-1, on HEV-like vessels may function to recruit CD8+ cytotoxic T cells in DSPTC. Additionally, sLeX-type glycans on carcinoma cells might partly contribute to highly metastatic properties of DSPTC through interaction with E-selectin expressed on endothelial cells.
Collapse
Affiliation(s)
- Shulin Low
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Yasuhiro Sakai
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Hitomi Hoshino
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | | | - Hiroto Kawashima
- Department of Microbiology and Molecular Genetics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Kayoko Higuchi
- Department of Diagnostic Pathology, Aizawa Hospital, Matsumoto, Japan
| | - Yoshiaki Imamura
- Division of Surgical Pathology, University of Fukui Hospital, Eiheiji, Japan
| | - Motohiro Kobayashi
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan.
| |
Collapse
|
22
|
Taga M, Hoshino H, Low S, Imamura Y, Ito H, Yokoyama O, Kobayashi M. A potential role for 6-sulfo sialyl Lewis X in metastasis of bladder urothelial carcinoma. Urol Oncol 2015; 33:496.e1-9. [PMID: 26137907 DOI: 10.1016/j.urolonc.2015.05.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/12/2015] [Accepted: 05/24/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVES It is widely accepted that sialyl Lewis X (sLeX) and sialyl Lewis A (sLeA, also known as CA 19-9) glycans expressed on cancer cells function in E-selectin-mediated metastasis. Recently, it was reported that 6-sulfo sLeX glycans detected by the MECA-79 monoclonal antibody are expressed in roughly a quarter of gastric adenocarcinoma cases, and that these cases show a poorer prognosis than MECA-79-negative cases do. The present study was undertaken to assess expression of 6-sulfo sLeX glycans in bladder urothelial carcinoma and evaluate potential clinical implications. MATERIALS AND METHODS We analyzed 78 specimens representing bladder urothelial carcinoma, as well as 4 bladder urothelial carcinoma cell lines, by immunostaining with a battery of anticarbohydrate antibodies. We also undertook an E-selectin·IgM chimera binding assay to assess E-selectin binding to 6-sulfo sLeX expressed on bladder urothelial carcinoma cells and performed reverse transcription polymerase chain reaction and complementary DNA transfection to determine which N-acetylglucosamine-6-O-sulfotransferases function in 6-sulfo sLeX biosynthesis in those cells. Finally, we performed double-immunofluorescence staining for MECA-79 and either CD3 or CD8 to evaluate potential association between high endothelial venule (HEV)-like vessels and tumor-infiltrating T lymphocytes. RESULTS 6-Sulfo sLeX glycans were expressed in ~20% of bladder urothelial carcinoma cases, particularly in plasmacytoid and micropapillary variants. Positive cells were also bound by E-selectin·IgM chimeras in a calcium-dependent manner. Transcripts encoding N-acetylglucosamine-6-O-sulfotransferase-2 were detected preferentially in HT-1197 bladder urothelial carcinoma cells expressing 6-sulfo sLeX, and transfection of the enzyme complementary DNA into HT-1376 cells, which do not express 6-sulfo sLeX glycans, resulted in cell surface expression of 6-sulfo sLeX. Furthermore, 6-sulfo sLeX glycans were expressed in HEV-like vessels induced in and around lymphocyte aggregates formed near carcinoma cell nests. These HEV-like vessel-associated tumor-infiltrating lymphocytes were composed primarily of CD3(+) T cells, with a fraction of CD8(+) cytotoxic T cells. CONCLUSIONS Our findings indicate that 6-sulfo sLeX glycans likely play 2 roles in bladder urothelial carcinoma progression: one in lymphocyte recruitment to enhance antitumor immune responses, and the other in E-selectin-mediated tumor cell adhesion to vascular endothelial cells, which is potentially associated with metastasis.
Collapse
Affiliation(s)
- Minekatsu Taga
- Division of Tumor Pathology, Department of Pathological Sciences, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan; Division of Urology, Department of Surgery, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Hitomi Hoshino
- Division of Tumor Pathology, Department of Pathological Sciences, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Shulin Low
- Division of Tumor Pathology, Department of Pathological Sciences, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Yoshiaki Imamura
- Division of Surgical Pathology, University of Fukui Hospital, Eiheiji, Japan
| | - Hideaki Ito
- Division of Urology, Department of Surgery, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Osamu Yokoyama
- Division of Urology, Department of Surgery, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Motohiro Kobayashi
- Division of Tumor Pathology, Department of Pathological Sciences, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan.
| |
Collapse
|
23
|
Peske JD, Woods AB, Engelhard VH. Control of CD8 T-Cell Infiltration into Tumors by Vasculature and Microenvironment. Adv Cancer Res 2015. [PMID: 26216636 DOI: 10.1016/bs.acr.2015.05.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CD8 T-cells are a critical brake on the initial development of tumors. In established tumors, the presence of CD8 T-cells is correlated with a positive patient prognosis, although immunosuppressive mechanisms limit their effectiveness and they are rarely curative without manipulation. Cancer immunotherapies aim to shift the balance back to dominant antitumor immunity through antibody blockade of immunosuppressive signaling pathways, vaccination, and adoptive transfer of activated or engineered T-cells. These approaches have yielded striking responses in small subsets of patients with solid tumors, most notably those with melanoma. Importantly, the subset of patients who respond to vaccination or immunosuppression blockade therapies are those with CD8 T-cells present in the tumor prior to initiating therapy. While current adoptive cell therapy approaches can be dramatically effective, they require infusion of extremely large numbers of T-cells, but the number that actually infiltrates the tumor is very small. Thus, poor representation of CD8 T-cells in tumors is a fundamental hurdle to successful immunotherapy, over and above the well-established barrier of immunosuppression. In this review, we discuss the factors that determine whether immune cells are present in tumors, with a focus on the representation of cytotoxic CD8 T-cells. We emphasize the critically important role of tumor-associated vasculature as a gateway that enables the active infiltration of both effector and naïve CD8 T-cells that exert antitumor activity. We also discuss strategies to enhance the gateway function and extend the effectiveness of immunotherapies to a broader set of cancer patients.
Collapse
Affiliation(s)
- J David Peske
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Amber B Woods
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Victor H Engelhard
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.
| |
Collapse
|
24
|
Sakai Y, Kobayashi M. Lymphocyte 'homing' and chronic inflammation. Pathol Int 2015; 65:344-54. [PMID: 25831975 DOI: 10.1111/pin.12294] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 03/04/2015] [Indexed: 12/20/2022]
Abstract
Chronic inflammation is a response to prolonged exposure to injurious stimuli that harm and destroy tissues and promote lymphocyte infiltration into inflamed sites. Following progressive accumulation of lymphocytes, the histology of inflamed tissue begins to resemble that of peripheral lymphoid organs, which can be referred to as lymphoid neogenesis or formation of tertiary lymphoid tissues. Lymphocyte recruitment to inflamed tissues is also reminiscent of lymphocyte homing to peripheral lymphoid organs. In the latter, under physiological conditions, homing receptors expressed on lymphocytes adhere to vascular addressin expressed on high endothelial venules (HEVs), initiating a lymphocyte migration process composed of sequential adhesive interactions. Intriguingly, in chronic inflammation, HEV-like vessels are induced de novo, despite the fact that the inflamed site is not originally lymphoid tissue, and these vessels contribute to lymphocyte recruitment in a manner similar to physiological lymphocyte homing. In this review, we first describe physiological lymphocyte homing mechanisms focusing on vascular addressins. We then describe HEV-like vessel-mediated pathogenesis seen in various chronic inflammatory disorders such as Helicobacter pylori gastritis, inflammatory bowel disease (IBD), autoimmune pancreatitis and sclerosing sialadenitis, as well as chronic inflammatory cell neoplasm MALT lymphoma, with reference to our work and that of others.
Collapse
Affiliation(s)
- Yasuhiro Sakai
- Division of Tumor Pathology, Department of Pathological Sciences, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Motohiro Kobayashi
- Division of Tumor Pathology, Department of Pathological Sciences, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| |
Collapse
|