1
|
Ding Z, Yang L, Wang X, Wang Y, Chen X. The expression and prognostic value of CCL19 in breast cancer. Discov Oncol 2025; 16:830. [PMID: 40392467 PMCID: PMC12092893 DOI: 10.1007/s12672-025-02715-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 05/14/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Breast cancer ranks as the foremost cause of cancer-related mortality among women globally. Timely diagnosis stands as the most effective approach in mitigating breast cancer mortality rates. There exists a close relationship between immune processes and tumorigenesis. This study aims to elucidate the immune mechanisms and potential biomarkers associated with breast cancer using bioinformatics techniques. OBJECTIVE Initially, differentially expressed genes were identified through consensus analysis of invasive breast cancer (BRCA) samples sourced from The Cancer Genome Atlas (TCGA) database, focusing on immunotherapy response. Subsequently, protein-protein interaction (PPI) networks, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were employed to refine the selection of potential genes. Lastly, expression and prognostic analyses of hub genes were conducted to identify reliable key genes, with a focus on CCL19. Immunohistochemistry (IHC) was employed to assess the differential expression of CCL19 in both tumor and adjacent breast tissue samples. Additionally, protein correlation analysis, signaling pathway analysis, immune infiltration analysis, gene co-expression analysis, and drug sensitivity analysis of CCL19 were conducted to investigate its pathological clinical features and potential biological functions. RESULTS CCL19 expression exhibited a significant increase in breast cancer. Elevated CCL19 expression correlates with advanced tumor stage and indicates a favorable prognosis in breast cancer. CCL19 expression correlates with the abundance of diverse tumor-infiltrating immune cells (TIICs). CCL19 exhibits a positive correlation with the expression of the majority of immune-related genes. Enrichment analysis revealed the involvement of CCL19 in immune-related pathways and the PI3K-Akt signaling pathway. These findings suggest that CCL19 may influence breast cancer prognosis through immune infiltration. Patients exhibiting high CCL19 expression demonstrated more favorable responses to immunotherapy. CONCLUSION Breast cancer demonstrates overexpression of CCL19. CCL19 holds promise as a biomarker for forecasting breast cancer prognosis and as a potential therapeutic target.
Collapse
Affiliation(s)
- Zonghao Ding
- General Surgery Department, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Yang
- AnHui Medical University, Hefei, China
| | | | - Yong Wang
- General Surgery Department, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiao Chen
- Thyroid and Breast Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Mughal SS, Reiss Y, Felsberg J, Meyer L, Macas J, Schlue S, Starzetz T, Köhrer K, Fehm T, Müller V, Lamszus K, Schadendorf D, Helfrich I, Wikman H, Berghoff A, Brors B, Plate KH, Reifenberger G. Identification and characterization of tertiary lymphoid structures in brain metastases. Acta Neuropathol Commun 2025; 13:91. [PMID: 40319321 PMCID: PMC12049775 DOI: 10.1186/s40478-025-02007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/13/2025] [Indexed: 05/07/2025] Open
Abstract
Brain metastases (BrM) are the most common cancers in the brain and linked to poor prognosis. Given the high incidence and often limited treatment options, understanding the complexity of the BrM tumor microenvironment is crucial for the development of novel therapeutic strategies. We performed transcriptome-wide gene expression profiling combined with spatial immune cell profiling to characterize the tumor immune microenvironment in 95 patients with BrM from different primary tumors. We found that BrM from lung carcinoma and malignant melanoma showed overall higher immune cell infiltration as compared to BrM from breast carcinoma. RNA sequencing-based immune cell deconvolution revealed gene expression signatures indicative of tertiary lymphoid structures (TLS) in subsets of BrM, mostly from lung cancer and melanoma. This finding was corroborated by multiplex immunofluorescence staining of immune cells in BrM tissue sections. Detection of TLS signatures was more common in treatment-naïve BrM and associated with prolonged survival after BrM diagnosis in lung cancer patients. Our findings highlight the cellular diversity of the tumor immune microenvironment in BrM of different cancer types and suggest a role of TLS formation for BrM patient outcome.
Collapse
Affiliation(s)
- Sadaf S Mughal
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120, Heidelberg, Germany.
| | - Yvonne Reiss
- Institute of Neurology (Edinger-Institute), University Hospital, Goethe University, Heinrich-Hoffmann-Strasse 7, 60590, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Paul-Ehrlich-Straße 42-44, 60596, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jörg Felsberg
- Institute of Neuropathology, Medical Faculty, Heinrich Heine University and University Hospital Düsseldorf, Moorenstrasse 5, Düsseldorf, Germany
| | - Lasse Meyer
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Jadranka Macas
- Institute of Neurology (Edinger-Institute), University Hospital, Goethe University, Heinrich-Hoffmann-Strasse 7, 60590, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Paul-Ehrlich-Straße 42-44, 60596, Frankfurt, Germany
| | - Silja Schlue
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Tatjana Starzetz
- Institute of Neurology (Edinger-Institute), University Hospital, Goethe University, Heinrich-Hoffmann-Strasse 7, 60590, Frankfurt, Germany
| | - Karl Köhrer
- Center for Biological and Medical Research (BMFZ), Genomics and Transcriptomics Laboratory (GTL), Heinrich Heine University, Universitätsstrasse 1, Düsseldorf, Germany
| | - Tanja Fehm
- Department of Gynecology and Obstetrics, Center of Integrated Oncology ABCD, Medical Faculty, Heinrich Heine University and University Hospital Düsseldorf, Moorenstrasse 5, Düsseldorf, Germany
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Katrin Lamszus
- Laboratory for Brain Tumor Biology, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 50, 45147, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Iris Helfrich
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 50, 45147, Essen, Germany
- Department of Dermatology and Allergy, University Hospital of Munich, Ludwig-Maximilian-University (LMU), Frauenlobstrasse 9-11, 80337, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Harriet Wikman
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Anna Berghoff
- Department of Internal Medicine 1, Clinical Division of Oncology, Medical University Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Medical Faculty and Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, 69120, Heidelberg, Germany
| | - Karl H Plate
- Institute of Neurology (Edinger-Institute), University Hospital, Goethe University, Heinrich-Hoffmann-Strasse 7, 60590, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Paul-Ehrlich-Straße 42-44, 60596, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Guido Reifenberger
- Institute of Neuropathology, Medical Faculty, Heinrich Heine University and University Hospital Düsseldorf, Moorenstrasse 5, Düsseldorf, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
3
|
Hameed H, Afzal M, Khan MA, Javaid L, Shahzad M, Abrar K. Unraveling the role of withanolides as key modulators in breast cancer mitigation. Mol Biol Rep 2025; 52:331. [PMID: 40117002 DOI: 10.1007/s11033-025-10442-1] [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: 01/21/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Addressing the elaborated landscape of therapeutics of global health concern i.e. breast cancer, this comprehensive review explores the promising effects of withanolides, bioactive compounds derived from Withania somnifera, for the treatment of breast cancer. In the breast, random mutations can accumulate over time, eventually transforming it into a tumor cell as certain receptors may be overexpressed by BC cells, which elicits downstream signaling and causes the production of genes involved in angiogenesis, survival, growth and migration, and other critical cell cycle practices. Merging insights from recent studies, our exploration delves into the molecular mechanisms that highlight withanolide's potential in the intervention of breast cancer. The study of apoptotic pathways unveils the withanolide's distinctive as well as pro-apoptotic effects, hinting at its effect as a potent modulator of the progression of breast cancer cells. Beyond its independent potential, there is a discussion on its distinctive perspective over the other therapies. Inweaving together these threads of evidence illuminates channels for future research. This review acts as a guide for researchers and clinicians negotiating the challenges of incorporating withanolides into the changing landscape for the treatment of breast cancer by balancing optimism with perceptive interpretation.
Collapse
Affiliation(s)
- Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Maham Afzal
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Mahtab Ahmad Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Laiba Javaid
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Maria Shahzad
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Kamran Abrar
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| |
Collapse
|
4
|
Takahashi H, Perez-Villarroel P, Falahat R, Mulé JJ. Targeting MARCO in combination with anti-CTLA-4 leads to enhanced melanoma regression and immune cell infiltration via macrophage reprogramming. J Immunother Cancer 2025; 13:e011030. [PMID: 40081947 PMCID: PMC11907082 DOI: 10.1136/jitc-2024-011030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Strategies to improve the therapeutic efficacy of cancer immunotherapy with immune checkpoint inhibitors include targeting additional immunosuppressive compartments in the tumor microenvironment (TME). Inhibitory macrophages (Mφ) can be one of the most abundant immune cells in the TME associated with poor prognosis. However, to date, selective Mφ depletion strategies as a cancer immunotherapy have not been successful in clinical trials. Macrophage Receptor with Collagenous Structure (MARCO) is one of a family of class-A scavenger receptors expressed by Mφ in the TME and is one of the most upregulated transcripts in dendritic cells (DC) following their ex vivo uptake of dead tumor cells. The clinical significance of MARCO expression in the TME is not fully understood. METHODS The therapeutic potential of targeting MARCO by an anti-murine MARCO (ED31, clone ED31) monoclonal antibody, which inhibits ligand-binding to MARCO, was explored in combination with anti-cytotoxic T-lymphocyte associated protein 4 (anti-CTLA-4) or anti-programmed cell death protein-1 (anti-PD-1) in C57BL/6J mice bearing B16F10 or Pan02 tumors. The mechanism by which ED31 impacts the TME was investigated by flow cytometry in the different treatment arms. The contribution of Mφ was assessed by both in vivo depletion and in vitro functional assays. Chemokine production was measured by a bead-based multiplex assay. RESULTS ED31 enhanced antitumor efficacy of anti-CTLA-4, but not of anti-PD-1. Analysis of the TME revealed that adding ED31 to anti-CTLA-4 substantially increased immune cell infiltration, including mature conventional DC recruitment, that was due to a switch to M1-pattern chemokines by Mφ. Mφ depletion completely abrogated both the increase in immune cell infiltration and chemokine production, and abolished the antitumor efficacy of the combination therapy. CONCLUSIONS Targeting MARCO as an additional checkpoint in the TME can offer a strategy to improve the antitumor efficacy of anti-CTLA-4 through a mechanism involving Mφ reprogramming rather than their depletion.
Collapse
Affiliation(s)
| | | | - Rana Falahat
- Immunology, Moffitt Cancer Center, Tampa, Florida, USA
| | - James J Mulé
- Immunology, Moffitt Cancer Center, Tampa, Florida, USA
- Cutaneous Oncology Program, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
5
|
Berglund AE, Puskas J, Yoder SJ, Smith AT, Marchion DC, Qin D, Mulé JJ, Torres-Roca JF, Eschrich SA. Evaluating the Radiation Sensitivity Index and 12-Chemokine Gene Expression Signature for Clinical Use in a CLIA Laboratory. CANCER RESEARCH COMMUNICATIONS 2025; 5:389-397. [PMID: 39932296 PMCID: PMC11873780 DOI: 10.1158/2767-9764.crc-24-0534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/22/2025] [Accepted: 02/06/2025] [Indexed: 02/19/2025]
Abstract
SIGNIFICANCE The RSI and 12CK GES are two GESs that predict tumor radiation sensitivity or the presence of tertiary lymphoid structures in tumors, respectively. These GESs were assessed within the CLIA process for future clinical use. We established proficiency, reproducibility, and reliability characteristics for both signatures in a controlled setting, indicating these GESs are suitable for validation within future clinical trials.
Collapse
Affiliation(s)
- Anders E. Berglund
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Quantitative Health Sciences, Mayo Clinic Florida, Jacksonville, Florida
| | - John Puskas
- Advanced Diagnostic Laboratory, Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Sean J. Yoder
- Molecular Genomics Core, Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Andrew T. Smith
- Molecular Genomics Core, Moffitt Cancer Center & Research Institute, Tampa, Florida
| | | | - Dahui Qin
- Advanced Diagnostic Laboratory, Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - James J. Mulé
- Department of Immunology, Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Javier F. Torres-Roca
- Department of Radiation Oncology, Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Steven A. Eschrich
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, Florida
| |
Collapse
|
6
|
Ribeiro V, Teillaud JL, Dieu-Nosjean MC, Lescaille G, Rochefort J. The prognostic significance of tertiary lymphoid structures in oral squamous cell carcinomas: a systematic review. FRONTIERS IN ORAL HEALTH 2025; 5:1524313. [PMID: 39911478 PMCID: PMC11794802 DOI: 10.3389/froh.2024.1524313] [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: 11/07/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
Introduction Upper aerodigestive tract cancers are prevalent, with a global incidence surpassing 500,000 new cases in 2018. Among these, oral squamous cell carcinomas (OSCC) constitute the majority. OSCC has a low 5-year survival rate due to late-stage diagnosis. Risk factors include alcohol and tobacco use. However, non-smokers and non-drinkers are also affected, especially young patients with tongue cancer. The impact of tumor microenvironment (TME) and tumor-infiltrating lymphocytes (TILs) on OSCC prognosis remains debated. Remarkably, Tertiary Lymphoid Structures (TLS) identified in solid tumors have shown associations with favorable outcomes, yet their prognostic significance in OSCC remains understudied. Objective Thus, this systematic review aims to explore the value of TLS in OSCC reported in the literature. Method A scoping review was conducted and six retrospective cohort studies involving 1,203 patients met the inclusion criteria. Results Predominantly male patients, with an average age of 49.3 years were included. Immunohistochemistry was the primary method to identify TLS, present in 21% up to 100% of cases. TLS were predominantly located in the peri-tumoral area (75.4%-84.8%) compared to the intra-tumoral area (33.8%-33.9%). Our review shows that the presence of TLS is associated with improved survival in OSCC. Discussion However, variations in TLS detection and classification methods across studies introduce potential biases, hindering direct comparisons between findings. For instance, reports that are based solely on examining HES-stained slides for TLS identification may raise reliability concerns. Standardization of methodologies is imperative to ensure consistency in criteria utilization, thereby facilitating meaningful data comparisons. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023428010, PROSPERO (CRD42023428010).
Collapse
Affiliation(s)
- V. Ribeiro
- Faculté de Santé, UFR Odontologie, Université Paris-Cité, Paris, France
| | - J-L. Teillaud
- UMRS 1135, Faculté de Santé Sorbonne Université, Sorbonne Université, Paris, France
- INSERM Unit 1135, Paris, France
- Laboratory “Immune Microenvironment and Immunotherapy”, Centre D’Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), Paris, France
| | - M-C. Dieu-Nosjean
- UMRS 1135, Faculté de Santé Sorbonne Université, Sorbonne Université, Paris, France
- INSERM Unit 1135, Paris, France
- Laboratory “Immune Microenvironment and Immunotherapy”, Centre D’Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), Paris, France
| | - G. Lescaille
- Faculté de Santé, UFR Odontologie, Université Paris-Cité, Paris, France
- UMRS 1135, Faculté de Santé Sorbonne Université, Sorbonne Université, Paris, France
- INSERM Unit 1135, Paris, France
- Laboratory “Immune Microenvironment and Immunotherapy”, Centre D’Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), Paris, France
- Service Odontologie, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - J. Rochefort
- Faculté de Santé, UFR Odontologie, Université Paris-Cité, Paris, France
- UMRS 1135, Faculté de Santé Sorbonne Université, Sorbonne Université, Paris, France
- INSERM Unit 1135, Paris, France
- Laboratory “Immune Microenvironment and Immunotherapy”, Centre D’Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), Paris, France
- Service Odontologie, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
7
|
Fan X, Feng D, Wei D, Li A, Wei F, Deng S, Shen M, Qin C, Yu Y, Liang L. Characterizing tertiary lymphoid structures associated single-cell atlas in breast cancer patients. Cancer Cell Int 2025; 25:12. [PMID: 39806382 PMCID: PMC11727541 DOI: 10.1186/s12935-025-03635-y] [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: 09/24/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
The tertiary lymphoid structure (TLS) is recognized as a potential prognosis factor for breast cancer and is strongly associated with response to immunotherapy. Inducing TLS neogenesis can enhance the immunogenicity of tumors and improve the efficacy of immunotherapy. However, our understanding of TLS associated region at the single-cell level remains limited. Therefore, we employed high-resolution techniques, including single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST), and a TLS-specific signature to investigate TLS associated regions in breast cancer. We identified eighteen cell types within the TLS associated regions and calculated differential expression genes by comparing TLS associated regions with other areas. Notably, macrophages in the TLS associated regions exhibit lineage transformation, shifting from facilitators of immune activation to supporters of tumor cell growth. In terms of cell-cell communication within the TLS associated regions, KRT86+ CD8+ T cells, HISTIH4C+ cycling CD8+ T cells, IFNG+ CD8+ T cells, and IGKV3-20+ B cells demonstrate strong interactions with other cells. Additionally, we found that APOD+ fibroblast and CCL21+ fibroblast primarily recruit T and B cells through the CXCL12-CXCR4 ligand-receptor signaling pathway. We also validate these findings in four independent breast cancer datasets, which include one cell-level resolution dataset from the 10 × Xenium platform and three spot-level datasets from the 10 × Visium platform.
Collapse
Affiliation(s)
- Xiaokai Fan
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China
| | - Daqin Feng
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Donggui Wei
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Anqi Li
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Fangyi Wei
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Shufang Deng
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Muling Shen
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Congzhi Qin
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Yongjia Yu
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China.
| | - Lun Liang
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China.
| |
Collapse
|
8
|
Peyraud F, Guegan JP, Vanhersecke L, Brunet M, Teyssonneau D, Palmieri LJ, Bessede A, Italiano A. Tertiary lymphoid structures and cancer immunotherapy: From bench to bedside. MED 2025; 6:100546. [PMID: 39798544 DOI: 10.1016/j.medj.2024.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 01/15/2025]
Abstract
Tertiary lymphoid structures (TLSs) are organized ectopic lymphoid aggregates within the tumor microenvironment that serve as crucial sites for the development of adaptive antitumor cellular and humoral immunity. TLSs have been consistently documented in numerous cancer types, correlating with improved prognosis and enhanced responses to immunotherapy, especially immune-checkpoint blockade (ICB). Given the potential role of TLSs as predictive biomarkers for the efficacy of ICB in cancer patients, the therapeutic manipulation of TLSs is gaining significant attention as a promising avenue for cancer treatment. Herein, we comprehensively review the composition, definition, and detection methods of TLSs in humans. We also discuss the contributions of TLSs to antitumor immunity, their prognostic value in cancer patients, and their association with therapeutic response to ICB-based immunotherapy. Finally, we present preclinical data supporting the potential of therapeutically manipulating TLSs as a promising approach for innovative cancer immunotherapy.
Collapse
Affiliation(s)
- Florent Peyraud
- Department of Medicine, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France; Explicyte Immuno-Oncology, Bordeaux, France.
| | | | - Lucile Vanhersecke
- Faculty of Medicine, University of Bordeaux, Bordeaux, France; Department of Pathology, Institut Bergonié, Bordeaux, France
| | - Maxime Brunet
- Department of Medicine, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - Diego Teyssonneau
- Department of Medicine, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France; Explicyte Immuno-Oncology, Bordeaux, France
| | - Lola-Jade Palmieri
- Department of Medicine, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France; Explicyte Immuno-Oncology, Bordeaux, France
| | | | - Antoine Italiano
- Department of Medicine, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France.
| |
Collapse
|
9
|
Yakushi A, Sugimoto M, Sasaki T. Co-expression network and survival analysis of breast cancer inflammation and immune system hallmark genes. Comput Biol Chem 2024; 113:108204. [PMID: 39270542 DOI: 10.1016/j.compbiolchem.2024.108204] [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: 04/04/2024] [Revised: 08/05/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024]
Abstract
The tertiary lymphoid structure (TLS) plays a central role in cancer immune response, and its gene expression pattern, called the TLS signature, has shown prognostic value in breast cancer. The formation of TLS and tumor-associated high endothelial venules (TA-HEVs), responsible for lymphocytic infiltration within the TLS, is associated with the expression of cancer hallmark genes (CHGs) related to immunity and inflammation. In this study, we performed co-expression network analysis of immune- and inflammation-related CHGs to identify predictive genes for breast cancer. In total, 382 immune- and inflammation-related CHGs with high expression variance were extracted from the GSE86166 microarray dataset of patients with breast cancer. CHGs were classified into five modules by applying weighted gene co-expression network analysis. The survival analysis results for each module showed that one module comprising 45 genes was statistically significant for relapse-free and overall survival. Four network properties identified key genes in this module with high prognostic prediction abilities: CD34, CXCL12, F2RL2, JAM2, PROS1, RAPGEF3, and SELP. The prognostic accuracy of the seven genes in breast cancer was synergistic and exceeded that of other predictors in both small and large public datasets. Enrichment analysis predicted that these genes had functions related to leukocyte infiltration of TA-HEVs. There was a positive correlation between key gene expression and the TLS signature, suggesting that gene expression levels are associated with TLS density. Co-expression network analysis of inflammation- and immune-related CHGs allowed us to identify genes that share a standard function in cancer immunity and have a high prognostic predictive value. This analytical approach may contribute to the identification of prognostic genes in TLS.
Collapse
Affiliation(s)
- Ayaka Yakushi
- Graduate School of Advanced Mathematical and Science, Meiji University, 4-21-1 Nakano, Nakano-ku, Tokyo, 164-8525, Japan
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan; Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Takanori Sasaki
- Graduate School of Advanced Mathematical and Science, Meiji University, 4-21-1 Nakano, Nakano-ku, Tokyo, 164-8525, Japan.
| |
Collapse
|
10
|
Campbell MJ, Wolf DM, Yau C, Brown-Swigart L, Wulfkuhle J, Gallagher IR, Zhu Z, Bolen J, Vandenberg S, Hoyt C, Mori H, Borowsky A, Sit L, Perlmutter J, Asare SM, Nanda R, Liu MC, Yee D, DeMichele AM, Hylton NM, Pusztai L, Berry DA, Hirst GL, Petricoin EF, Veer LV, Esserman L. Multi-platform biomarkers of response to an immune checkpoint inhibitor in the neoadjuvant I-SPY 2 trial for early-stage breast cancer. Cell Rep Med 2024; 5:101799. [PMID: 39510069 PMCID: PMC11604542 DOI: 10.1016/j.xcrm.2024.101799] [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: 07/25/2022] [Revised: 05/13/2024] [Accepted: 09/29/2024] [Indexed: 11/15/2024]
Abstract
Only a subset of patients with breast cancer responds to immune checkpoint blockade (ICB). To better understand the underlying mechanisms, we analyze pretreatment biopsies from patients in the I-SPY 2 trial who receive neoadjuvant ICB using multiple platforms to profile the tumor microenvironment. A variety of immune cell populations and markers of immune/cytokine signaling associate with pathologic complete response (pCR). Interestingly, these differ by breast cancer receptor subtype. Measures of the spatial distributions of immune cells within the tumor microenvironment, in particular colocalization or close spatial proximity of PD-1+ T cells with PD-L1+ cells (immune and tumor cells), are significantly associated with response in the overall cohort as well as the in the triple negative (TN) and HR+HER2- subtypes. Our findings indicate that biomarkers associated with immune cell signaling, immune cell densities, and spatial metrics are predictive of neoadjuvant ICB efficacy in breast cancer.
Collapse
Affiliation(s)
- Michael J Campbell
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Denise M Wolf
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lamorna Brown-Swigart
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Julie Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Isela R Gallagher
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Zelos Zhu
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jennifer Bolen
- Biospecimen Resource Program (BIOS), University of California, San Francisco, San Francisco, CA 94143, USA
| | - Scott Vandenberg
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Hidetoshi Mori
- Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA 95616, USA
| | - Alexander Borowsky
- Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA 95616, USA; Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Laura Sit
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Smita M Asare
- Quantum Leap Healthcare Collaborative, San Francisco, CA 94118, USA
| | - Rita Nanda
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Minetta C Liu
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Douglas Yee
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Angela M DeMichele
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nola M Hylton
- Department of Radiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lajos Pusztai
- Yale School of Medicine, Yale University, New Haven, CT 06510, USA
| | | | - Gillian L Hirst
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Laura Van't Veer
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
11
|
Yu C, Xu J, Xu S, Tang L, Han Q, Sun Z. Research trends, hotspots and future directions of tertiary lymphoid structures in cancer: a comprehensive informatics analysis and visualization study. Discov Oncol 2024; 15:665. [PMID: 39549226 PMCID: PMC11569082 DOI: 10.1007/s12672-024-01556-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Abstract
Many studies have reported the presence of tertiary lymphoid structures (TLSs) in cancer, but the research progress of TLSs in cancer has not been systematically analyzed. Therefore, we analyzed the global scientific knowledge in the field using informatics methods. The results showed that TLSs in cancer have received increasing attention since the 21st century, with an annual publication growth rate of 27.86%. Unsupervised hierarchical clustering based on machine learning further categorized the research features into four clusters, with the cluster related to immunotherapy being considered an emerging cluster. TLSs and immunotherapy were identified as the top two hotspots with the highest occurrence frequency and total link strength. The Walktrap algorithm indicated that "TLSs, carcinoma, prognostic value" and "high endothelial venules, germinal-centers, node-like structures" are important to TLSs but remain underexplored, representing promising research directions. These findings suggest that cancer-related TLSs have brought new insights into antitumor immunity, and targeting TLSs has the potential to transform the landscape of antitumor immunotherapy.
Collapse
Affiliation(s)
- Chengdong Yu
- Jiangxi Medical College, Nanchang University, Nanchang, China
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, China
| | - Jiawei Xu
- Jiangxi Medical College, Nanchang University, Nanchang, China
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, China
| | - Siyi Xu
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, China
| | - Lei Tang
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, China
| | - Qinyuan Han
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhengkui Sun
- Jiangxi Medical College, Nanchang University, Nanchang, China.
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, China.
| |
Collapse
|
12
|
Deng S, Yang X, He L, Hou Y, Meng H. Tertiary Lymphoid Structures in Microorganism-Related Cancer. Cancers (Basel) 2024; 16:3464. [PMID: 39456558 PMCID: PMC11505735 DOI: 10.3390/cancers16203464] [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: 09/04/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) are ectopic lymphoid tissues formed by the accumulation of lymphocytes and other components outside lymphoid organs. They have been shown to be widespread in cancers and have predictive effects on prognosis and immunotherapy efficacy; however, there is no standardized measurement guide. This paper provides a reference for future research. Moreover, the induction strategy for the formation mechanism of TLSs is a new direction for future cancer treatment, such as cancer vaccines for microorganisms. The effects of microorganisms on cancer are dual. The role of microorganisms, including bacteria, parasites, viruses, and fungi, in promoting cancer has been widely confirmed. However, the specific mechanism of their tumor suppressor effect, particularly the promotion of TLS formation, is currently unknown. In this review, we summarize the role of TLSs in cancer related to microbial infection and provide new ideas for further understanding their mechanisms of action in cancer.
Collapse
Affiliation(s)
- Shuzhe Deng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150086, China;
| | - Xinxin Yang
- Precision Medical Center, Harbin Medical University Cancer Hospital, Harbin 150086, China; (X.Y.); (Y.H.)
| | - Lin He
- Department of Stomatology, Heilongjiang Provincial Hospital, Harbin 150000, China;
| | - Yunjing Hou
- Precision Medical Center, Harbin Medical University Cancer Hospital, Harbin 150086, China; (X.Y.); (Y.H.)
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150086, China;
- Precision Medical Center, Harbin Medical University Cancer Hospital, Harbin 150086, China; (X.Y.); (Y.H.)
| |
Collapse
|
13
|
Berglund A, Puskas J, Yoder S, Smith AT, Marchion DC, Qian D, Mulé JJ, Torres-Roca JF, Eschrich SA. Evaluating the Radiation Sensitivity Index and 12-chemokine gene expression signature for clinical use in a CLIA laboratory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613957. [PMID: 39345465 PMCID: PMC11429982 DOI: 10.1101/2024.09.19.613957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Background The radiation sensitivity index (RSI) and 12-chemokine gene expression signature (12CK GES) are two gene expression signatures (GES) that were previously developed to predict tumor radiation sensitivity or identify the presence of tertiary lymphoid structures in tumors, respectively. To advance the use of these GES into clinical trial evaluation, their assays must be assessed within the context of the Clinical Laboratory Improvement Amendments (CLIA) process. Methods Using HG-U133Plus 2.0 arrays, we first established CLIA laboratory proficiency. Then the accuracy (limit of detection and macrodissection impact), precision (variability by time and operator), sample type (surgery vs. biopsy), and concordance with reference laboratory were evaluated. Results RSI and 12CK GES were reproducible (RSI: 0.01 mean difference, 12CK GES 0.17 mean difference) and precise with respect to time and operator. Taken together, the reproducibility analysis of the scores indicated a median RSI difference of 0.06 (6.47% of range) across samples and a median 12CK GES difference of 0.92 (12.29% of range). Experiments indicated that the lower limit of input RNA is 5 ng. Reproducibility with a second CLIA laboratory demonstrated reliability with the median RSI score difference of 0.065 (6% of full range) and 12CK GES difference of 0.93 (12 % of observed range). Conclusions Overall, under CLIA, RSI and 12CK GES were demonstrated by the Moffitt Cancer Center Advanced Diagnostic Laboratory to be reproducible GES for clinical usage.
Collapse
Affiliation(s)
- Anders Berglund
- Department of Biostatistics & Bioinformatics, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, 33612, FL, USA
| | - John Puskas
- Advanced Diagnostic Laboratory, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, 33612, FL, USA
| | - Sean Yoder
- Molecular Genomics Core, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, 33612, FL, USA
| | - Andrew T. Smith
- Molecular Genomics Core, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, 33612, FL, USA
| | - Douglas C. Marchion
- Tissue Core, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, 33612, FL, USA
| | - Dahui Qian
- Advanced Diagnostic Laboratory, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, 33612, FL, USA
| | - James J. Mulé
- Department of Immunology, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, 33612, FL, USA
| | - Javier F. Torres-Roca
- Department of Radiation Oncology, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, 33612, FL, USA
| | - Steven A. Eschrich
- Department of Biostatistics & Bioinformatics, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, 33612, FL, USA
| |
Collapse
|
14
|
Ma Y, Li X, Zhang J, Zhao X, Lu Y, Shen G, Wang G, Liu H, Hao J. Integrating tertiary lymphoid structure-associated genes into computational models to evaluate prognostication and immune infiltration in pancreatic cancer. J Leukoc Biol 2024; 116:589-600. [PMID: 38484172 DOI: 10.1093/jleuko/qiae067] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/03/2024] [Accepted: 02/27/2024] [Indexed: 09/03/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by poor response to all therapeutic modalities and dismal prognosis. The presence of tertiary lymphoid structures (TLSs) in various solid cancers is of crucial prognostic significance, highlighting the intricate interplay between the tumor microenvironment and immune cells aggregation. However, the extent to which TLSs and immune status affect PDAC prognosis remains incompletely understood. Here, we sought to unveil the unique properties of TLSs in PDAC by leveraging both single-cell and bulk transcriptomics, culminating in a risk model that predicts clinical outcomes. We used TLS scores based on a 12-gene (CCL2, CCL3, CCL4, CCL5, CCL8, CCL18, CCL19, CCL21, CXCL9, CXCL10, CXCL11, and CXCL13) and 9-gene (PTGDS, RBP5, EIF1AY, CETP, SKAP1, LAT, CCR6, CD1D, and CD79B) signature, respectively, and examined their distribution in cell clusters of single-cell data from PDAC samples. The markers involved in these clusters were selected to develop a prognostic model using The Cancer Genome Atlas Program database as the training cohort and Gene Expression Omnibus database as the validation cohort. Further, we compared the immune infiltration, drug sensitivity, and enriched and differentially expressed genes between the high- and low-risk groups in our model. Therefore, we established a risk model that has significant implications for the prognostic assessment of PADC patients with remarkable differences in immune infiltration and chemosensitivity between the low- and high-risk groups. This paradigm established by TLS-related cell marker genes provides a prognostic prediction and a panel of novel therapeutic targets for exploring potential immunotherapy.
Collapse
Affiliation(s)
- Ying Ma
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Xuesong Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Jin Zhang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Xiangqin Zhao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Yi Lu
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Guangcong Shen
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Guowen Wang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Hong Liu
- Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, West Huanhu Road, Hexi District, Tianjin 300060, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, West Huanhu Road, Hexi District, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Jihui Hao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| |
Collapse
|
15
|
Niu L, Chen T, Yang A, Yan X, Jin F, Zheng A, Song X. Macrophages and tertiary lymphoid structures as indicators of prognosis and therapeutic response in cancer patients. Biochim Biophys Acta Rev Cancer 2024; 1879:189125. [PMID: 38851437 DOI: 10.1016/j.bbcan.2024.189125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/24/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Tertiary lymphoid structures (TLS) can reflect cancer prognosis and clinical outcomes in various tumour tissues. Tumour-associated macrophages (TAMs) are indispensable components of the tumour microenvironment and play crucial roles in tumour development and immunotherapy. TAMs are associated with TLS induction via the modulation of the T cell response, which is a major component of the TLS. Despite their important roles in cancer immunology, the subtypes of TAMs that influence TLS and their correlation with prognosis are not completely understood. Here, we provide novel insights into the role of TAMs in regulating TLS formation. Furthermore, we discuss the prognostic value of these TAM subtypes and TLS, as well as the current antitumour therapies for inducing TLS. This study highlights an entirely new field of TLS regulation that may lead to the development of an innovative perspective on immunotherapy for cancer treatment.
Collapse
Affiliation(s)
- Li Niu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ting Chen
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Aodan Yang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China
| | - Xiwen Yan
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China
| | - Feng Jin
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China
| | - Ang Zheng
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China.
| | - Xinyue Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.
| |
Collapse
|
16
|
Jung H, Paust S. Chemokines in the tumor microenvironment: implications for lung cancer and immunotherapy. Front Immunol 2024; 15:1443366. [PMID: 39114657 PMCID: PMC11304008 DOI: 10.3389/fimmu.2024.1443366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
The tumor microenvironment (TME) is a complex interconnected network of immune cells, fibroblasts, blood vessels, and extracellular matrix surrounding the tumor. Because of its immunosuppressive nature, the TME can pose a challenge for cancer immunotherapies targeting solid tumors. Chemokines have emerged as a crucial element in enhancing the efficacy of cancer immunotherapy, playing a direct role in immune cell signaling within the TME and facilitating immune cell migration towards cancer cells. However, chemokine ligands and their receptors exhibit context-dependent diversity, necessitating evaluation of their tumor-promoting or inhibitory effects based on tumor type and immune cell characteristics. This review explores the role of chemokines in tumor immunity and metastasis in the context of the TME. We also discuss current chemokine-related advances in cancer immunotherapy research, with a particular focus on lung cancer, a common cancer with a low survival rate and limited immunotherapy options.
Collapse
Affiliation(s)
| | - Silke Paust
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| |
Collapse
|
17
|
Weng Y, Yuan J, Cui X, Wang J, Chen H, Xu L, Chen X, Peng M, Song Q. The impact of tertiary lymphoid structures on tumor prognosis and the immune microenvironment in non-small cell lung cancer. Sci Rep 2024; 14:16246. [PMID: 39009684 PMCID: PMC11250816 DOI: 10.1038/s41598-024-64980-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] [Received: 02/18/2024] [Accepted: 06/14/2024] [Indexed: 07/17/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is a common malignancy whose prognosis and treatment outcome are influenced by many factors. Some studies have found that tertiary lymphoid structures (TLSs) in cancer may contribute to prognosis and the prediction of immunotherapy efficacy However, the combined role of TLSs in NSCLC remains unclear. We accessed The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases to obtain mRNA sequencing data and clinical information as the TCGA cohort, and used our own sample of 53 advanced NSCLC as a study cohort. The samples were divided into TLS+ and TLS- groups by pathological tissue sections. Patients of the TLS+ group had a better OS (p = 0.022), PFS (p = 0.042), and DSS (p = 0.004) in the TCGA cohort, and the results were confirmed by the study cohort (PFS, p = 0.012). Furthermore, our result showed that the count and size of TLSs are closely associated with the efficacy of immunotherapy. In addition, the TLS+ group was associated with better immune status and lower tumor mutation load. In the tumor microenvironment (TME), the expression levels of CD4+ T cells and CD8+ T cells of different phenotypes were associated with TLSs. Overall, TLSs are a strong predictor of survival and immunotherapeutic efficacy in advanced NSCLC, and T cell-rich TLSs suggest a more ordered and active immune response site, which aids in the decision-making and application of immunotherapy in the clinic.
Collapse
Affiliation(s)
- Yiming Weng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xue Cui
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jinsong Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Honglei Chen
- Department of Pathology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Li Xu
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinyi Chen
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Min Peng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Qibin Song
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
18
|
Mei SQ, Liu JQ, Huang ZJ, Luo WC, Peng YL, Chen ZH, Deng Y, Xu CR, Zhou Q. Identification of a risk score model based on tertiary lymphoid structure-related genes for predicting immunotherapy efficacy in non-small cell lung cancer. Thorac Cancer 2024; 15:1119-1131. [PMID: 38558529 DOI: 10.1111/1759-7714.15299] [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: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Tertiary lymphoid structures (TLSs) affect the prognosis and efficacy of immunotherapy in patients with non-small cell lung cancer (NSCLC), but the underlying mechanisms are not well understood. METHODS TLSs were identified and categorized online from the Cancer Digital Slide Archive (CDSA). Overall survival (OS) and disease-free survival (DFS) were analyzed. GSE111414 and GSE136961 datasets were downloaded from the GEO database. GSVA, GO and KEGG were used to explore the signaling pathways. Immune cell infiltration was analyzed by xCell, ssGSEA and MCP-counter. The analysis of WGCNA, Lasso and multivariate cox regression were conducted to develop a gene risk score model based on the SU2C-MARK cohort. RESULTS TLS-positive was a protective factor for OS according to multivariate cox regression analysis (p = 0.029). Both the TLS-positive and TLS-mature groups exhibited genes enrichment in immune activation pathways. The TLS-mature group showed more activated dendritic cell infiltration than the TLS-immature group. We screened TLS-related genes using WGCNA. Lasso and multivariate cox regression analysis were used to construct a five-genes (RGS8, RUF4, HLA-DQB2, THEMIS, and TRBV12-5) risk score model, the progression free survival (PFS) and OS of patients in the low-risk group were markedly superior to those in the high-risk group (p < 0.0001; p = 0.0015, respectively). Calibration and ROC curves indicated that the combined model with gene risk score and clinical features could predict the PFS of patients who have received immunotherapy more accurately than a single clinical factor. CONCLUSIONS Our data suggested a pivotal role of TLSs formation in survival outcome and immunotherapy response of NSCLC patients. Tumors with mature TLS formation showed more activated immune microenvironment. In addition, the model constructed by TLS-related genes could predict the response to immunotherapy and is meaningful for clinical decision-making.
Collapse
Affiliation(s)
- Shi-Qi Mei
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jia-Qi Liu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zi-Jian Huang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wei-Chi Luo
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ying-Long Peng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine South China University of Technology, Guangzhou, China
| | - Zhi-Hong Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yu Deng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chong-Rui Xu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
19
|
Xiao Y, Liu Y, Sun Y, Huang C, Zhong S. MEIS2 suppresses breast cancer development by downregulating IL10. Cancer Rep (Hoboken) 2024; 7:e2064. [PMID: 38711262 PMCID: PMC11074520 DOI: 10.1002/cnr2.2064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/06/2024] [Accepted: 03/23/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is the most commonly diagnosed female cancer. Homeobox protein MEIS2, a key transcription factor, is involved in the regulation of many developmental and cellular processes. However, the role of MEIS2 in the development of breast cancer is still unclear. AIMS We aimed to examine the role of myeloid ecotropic insertion site (MEIS2) in breast cancer and the association of MEIS2 with breast cancer clinical stages and pathological grades. We revealed the underlying mechanism by which MEIS2 affected breast cancer cell growth and tumor development. METHODS AND RESULTS Using human BC cell lines, clinical samples and animal xenograft model, we reveal that MEIS2 functions as a tumor suppressor in breast cancer. The expression of MEIS2 is inversely correlated with BC clinical stages and pathological grades. MEIS2 knockdown (MEIS2-KD) promotes while MEIS2 overexpression suppresses breast cancer cell proliferation and tumor development in vitro and in animal xenograft models, respectively. To determine the biological function of MEIS2, we screen the expression of a group of MEIS2 potential targeting genes in stable-established cell lines. Results show that the knockdown of MEIS2 in breast cancer cells up-regulates the IL10 expression, but MEIS2 overexpression opposed the effect on IL10 expression. Furthermore, the suppressive role of MEIS2 in breast cancer cell proliferation is associated with the IL10 expression and myeloid cells infiltration. CONCLUSION Our study demonstrates that the tumor suppressor of MEIS2 in breast cancer progression is partially via down regulating the expression of IL10 and promoting myeloid cells infiltration. Targeting MEIS2 would be a potentially therapeutic avenue for BC.
Collapse
Affiliation(s)
- Yongzhi Xiao
- Department of Ultrasound Diagnosis, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yingzhe Liu
- Xiangya International Medical Center, National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yangqing Sun
- Department of Oncology, Xiangya HospitalCentral South UniversityHunanChina
| | - Changhao Huang
- Department of Oncology, Xiangya HospitalCentral South UniversityHunanChina
| | - Shangwei Zhong
- The Cancer Research Institute, Hengyang Medical SchoolUniversity of South ChinaHengyangChina
| |
Collapse
|
20
|
Li S, Wang Z, Huang HD, Lee TY. Machine Learning-Based Characterization and Identification of Tertiary Lymphoid Structures Using Spatial Transcriptomics Data. Int J Mol Sci 2024; 25:3887. [PMID: 38612697 PMCID: PMC11011734 DOI: 10.3390/ijms25073887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/22/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) are organized aggregates of immune cells in non-lymphoid tissues and are associated with a favorable prognosis in tumors. However, TLS markers remain inconsistent, and the utilization of machine learning techniques for this purpose is limited. To tackle this challenge, we began by identifying TLS markers through bioinformatics analysis and machine learning techniques. Subsequently, we leveraged spatial transcriptomic data from Gene Expression Omnibus (GEO) and built two support vector classifier models for TLS prediction: one without feature selection and the other using the marker genes. The comparable performances of these two models confirm the efficacy of the selected markers. The majority of the markers are immunoglobulin genes, demonstrating their importance in the identification of TLSs. Our research has identified the markers of TLSs using machine learning methods and constructed a model to predict TLS location, contributing to the detection of TLS and holding the promising potential to impact cancer treatment strategies.
Collapse
Affiliation(s)
- Songyun Li
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (S.L.); (Z.W.)
| | - Zhuo Wang
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (S.L.); (Z.W.)
| | - Hsien-Da Huang
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (S.L.); (Z.W.)
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| |
Collapse
|
21
|
Zhao H, Zhao Y, Zhang S, Wang Z, Yu W, Dong N, Yang X, Zhang X, Sun Q, Hao X, Ren X. Effects of immunogenic cell death-inducing chemotherapeutics on the immune cell activation and tertiary lymphoid structure formation in melanoma. Front Immunol 2024; 15:1302751. [PMID: 38384466 PMCID: PMC10879401 DOI: 10.3389/fimmu.2024.1302751] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
Background The infiltration and activation of immune cells in the tumor microenvironment (TIME) affect the prognosis of patients with cancer. Tertiary lymphoid structure (TLS) formation favors tumour- infiltrating-lymphocyte (TIL) recruitment and is regarded as an important indicator of good prognosis associated with immunotherapy in patients with tumors. Chemotherapy is currently one of the most commonly used clinical treatment methods. However, there have been no clear report to explore the effects of different types of chemotherapy on TLS formation in the TIME. This study examined the effects of immunogenic cell death (ICD)-inducing chemotherapeutics on immune cells, high-endothelial venules (HEV), and TLSs in mouse melanomas. Methods Doxorubicin (an ICD inducer), gemcitabine (non-ICD inducer), and a combination of the two drugs was delivered intra-peritoneally to B16F1-loaded C57BL/6 mice. The infiltration of immune cells into tumor tissues was evaluated using flow cytometry. HEV and TLS formation was assessed using immunohistochemistry and multiple fluorescent immunohistochemical staining. Results Doxorubicin alone, gemcitabine alone, and the two-drug combination all slowed tumor growth, with the combined treatment demonstrating a more pronounced effect. Compared with the control group, the doxorubicin group showed a higher infiltration of CD8+ T cells and tissue-resident memory T cells (TRM) and an increase in the secretion of interferon-γ, granzyme B, and perforin in CD8+ T subsets and activation of B cells and dendritic cells. Doxorubicin alone and in combination with gemcitabine decreased regulatory T cells in the TIME. Moreover, doxorubicin treatment promoted the formation of HEV and TLS. Doxorubicin treatment also upregulated the expression of programmed cell death protein (PD)-1 in CD8+ T cells and programmed cell death protein ligand (PD-L)1 in tumor cells. Conclusions These results indicate that doxorubicin with an ICD reaction promotes TLS formation and increases PD-1/PD-L1 expression in tumor tissues. The results demonstrate the development of a therapeutic avenue using combined immune checkpoint therapy.
Collapse
Affiliation(s)
- Hua Zhao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Haihe Laboratory of Cell Ecosystem, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yu Zhao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Siyuan Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhe Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenwen Yu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Nan Dong
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xuena Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiying Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qian Sun
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Haihe Laboratory of Cell Ecosystem, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xishan Hao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Haihe Laboratory of Cell Ecosystem, Tianjin, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Haihe Laboratory of Cell Ecosystem, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
22
|
Gao Z, Wan Z, Yu P, Shang Y, Zhu G, Jiang H, Chen Y, Wang S, Lei F, Huang W, Zeng Q, Wang Y, Rong W, Hong Y, Gao Q, Niu P, Zhai Z, An K, Ding C, Wang Y, Gu G, Wang X, Meng Q, Ye S, Liu H, Gu J. A recurrence-predictive model based on eight genes and tumor mutational burden/microsatellite instability status in Stage II/III colorectal cancer. Cancer Med 2024; 13:e6720. [PMID: 38111983 PMCID: PMC10807589 DOI: 10.1002/cam4.6720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/18/2023] [Accepted: 10/27/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Although adjuvant chemotherapy (ACT) is widely used to treat patients with Stage II/III colorectal cancer (CRC), administering ACT to specific patients remains a challenge. The decision to ACT requires an accurate assessment of recurrence risk and absolute treatment benefit. However, the traditional TNM staging system does not accurately assess a patient's individual risk of recurrence. METHODS To identify recurrence risk-related genetic factors for Stage II/III CRC patients after radical surgery, we conducted an analysis of whole-exome sequencing of 47 patients with Stage II/III CRC who underwent radical surgery at five institutions. Patients were grouped into non-recurrence group (NR, n = 24, recurrence-free survival [RFS] > 5 years) and recurrence group (R, n = 23, RFS <2 years). The TCGA-COAD/READ cohort was employed as the validation dataset. RESULTS A recurrence-predictive model (G8plus score) based on eight gene (CUL9, PCDHA12, HECTD3, DCX, SMARCA2, FAM193A, AATK, and SORCS2) mutations and tumor mutation burden/microsatellite instability (TMB/MSI) status was constructed, with 97.87% accuracy in our data and 100% negative predictive value in the TCGA-COAD/READ cohort. For the TCGA-COAD/READ cohort, the G8plus-high group had better RFS (HR = 0.22, p = 0.024); the G8plus-high tumors had significantly more infiltrated immune cell types, higher tertiary lymphoid structure signature scores, and higher immunological signature scores. The G8plus score was also a predict biomarker for immunotherapeutic in advanced CRC in the PUCH cohort. CONCLUSIONS In conclusion, the G8plus score is a powerful biomarker for predicting the risk of recurrence in patients with stage II/III CRC. It can be used to stratify patients who benefit from ACT and immunotherapy.
Collapse
Affiliation(s)
- Zhaoya Gao
- Department of General SurgeryPeking University First HospitalBeijingChina
| | - Zhiyi Wan
- Genecast Biotechnology Co., Ltd.Wuxi CityJiangsu ProvinceChina
| | - Pengfei Yu
- Department of General SurgeryAir Force Medical Center, Chinese People's Liberation ArmyBeijingChina
| | - Yan Shang
- Department of Colorectal SurgeryCancer Hospital of China Medical University, Liaoning Cancer Hospital and InstituteShenyangLiaoning ProvinceChina
| | - Guangsheng Zhu
- Department of Gastrointestinal SurgeryHubei Cancer HospitalWuhanHubei ProvinceChina
| | - Huiyuan Jiang
- Department of Colorectal and Anal SurgeryShanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanShanxi ProvinceChina
| | - Yawei Chen
- Genecast Biotechnology Co., Ltd.Wuxi CityJiangsu ProvinceChina
| | - Shengzhou Wang
- Genecast Biotechnology Co., Ltd.Wuxi CityJiangsu ProvinceChina
| | - Fuming Lei
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
| | - Wensheng Huang
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
| | - Qingmin Zeng
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
| | - Yanzhao Wang
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
| | - Wanshui Rong
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
| | - Yuming Hong
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
| | - Qingkun Gao
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
| | - Pengfei Niu
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
| | - Zhichao Zhai
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
| | - Ke An
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
| | - Changmin Ding
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
| | - Yunfan Wang
- Department of PathologyPeking University Shougang HospitalBeijingChina
| | - Guoli Gu
- Department of General SurgeryAir Force Medical Center, Chinese People's Liberation ArmyBeijingChina
| | - Xin Wang
- Department of General SurgeryPeking University First HospitalBeijingChina
| | - Qingkai Meng
- Department of Colorectal SurgeryCancer Hospital of China Medical University, Liaoning Cancer Hospital and InstituteShenyangLiaoning ProvinceChina
| | - Shengwei Ye
- Department of Gastrointestinal SurgeryHubei Cancer HospitalWuhanHubei ProvinceChina
| | - Haiyi Liu
- Department of Colorectal and Anal SurgeryShanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanShanxi ProvinceChina
| | - Jin Gu
- Department of Gastrointestinal SurgeryPeking University Shougang HospitalBeijingChina
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal SurgeryPeking University Cancer Hospital & InstituteBeijingChina
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
- Peking University International Cancer InstituteBeijingChina
| |
Collapse
|
23
|
Lin X, Zhao X, Chen Y, Yang R, Dai Z, Li W, Lin C, Cao W. CXC ligand 13 orchestrates an immunoactive microenvironment and enhances immunotherapy response in head and neck squamous cell carcinoma. Int J Immunopathol Pharmacol 2024; 38:3946320241227312. [PMID: 38252495 PMCID: PMC10807398 DOI: 10.1177/03946320241227312] [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: 06/15/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Objectives: This study aims to systematically explore the role of chemokine CXC ligand 13 (CXCL13) in head and neck squamous cell carcinoma (HNSCC). Methods: The Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) databases provided the RNA-seq data for cancer and normal tissues, respectively. Gene set enrichment analysis was applied to search the cancer hallmarks associated with CXCL13 expression. TIMER2.0 was the main platform used to investigate the immune cell infiltration related to CXCL13. Immunohistochemistry was applied to explore the relationship between CXCL13 and patients' prognosis and the relationship between CXCL13 and tertiary lymphoid structures (TLSs). Results: The expression of CXCL13 was upregulated in most tumors, including HNSCC. The higher expression of CXCL13 was closely related to the positive prognosis of HNSCC. CXCL13 was mainly expressed in B cells and CD8 + T cells, revealing the relationship between its expression and immune activation in the tumor microenvironment. Furthermore, immunohistochemistry and multiple fluorescence staining analysis of HNSCC samples showed a powerful correlation between CXCL13 expression, TLSs formation, and positive prognosis. Finally, CXCL13 significantly increased the response to cancer immunotherapy. Conclusions: CXCL13 may function as a potential biomarker for predicting prognosis and immunotherapy response and associate with TLSs in HNSCC.
Collapse
Affiliation(s)
- Xiaohu Lin
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Xiaomei Zhao
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yiming Chen
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Rong Yang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhenlin Dai
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Wei Li
- Xuchang Central Hospital, Henan, China
| | - Chengzhong Lin
- Department of Oral Maxillofacial Surgery, Zhongshan Hospital, Fu Dan University, Shanghai, China
| | - Wei Cao
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
24
|
Wu Y, Zhao J, Wang Z, Liu D, Tian C, Ye B, Sun Y, Li H, Wang X. Association of systemic inflammatory markers and tertiary lymphoid structure with pathological complete response in gastric cancer patients receiving preoperative treatment: a retrospective cohort study. Int J Surg 2023; 109:4151-4161. [PMID: 38259000 PMCID: PMC10720847 DOI: 10.1097/js9.0000000000000741] [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: 06/21/2023] [Accepted: 08/24/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND Assessment of systemic and local immune responses is crucial in determining the efficacy of cancer interventions. The identification of specific factors that correlate with pathological complete response (pCR) is essential for optimizing treatment decisions. METHODS In this retrospective study, a total of 521 patients diagnosed with gastric adenocarcinoma who underwent curative gastrectomy following preoperative treatment were reviewed. Of these patients, 463 did not achieve pCR (non-pCR) and 58 achieved pCR. Clinicopathological factors were evaluated to identify predictors for pCR using a logistic regression model. Additionally, a smaller cohort (n=76) was derived using propensity score matching to investigate local immune response, specifically the features of tertiary lymphoid structure (TLS) using H&E staining, immunohistochemistry, and multiplex immunofluorescence. RESULTS The multivariate regression analysis demonstrated a significant association between low systemic inflammatory status and pCR, as evidenced by reduced levels of the combined systemic immune-inflammation index (SII) and neutrophil-to-lymphocyte ratio (NLR) (SII+NLR) (odds ratio: 3.33, 95% CI: 1.79-6.17, P<0.001). In the smaller cohort analysis, distinct TLS characteristics were correlated with the presence of pCR. Specifically, a higher density of TLS and a lower proportion of PD1+ cells and CD8+ cells within TLS in the tumor bed were strongly associated with pCR. CONCLUSION Both systemic and local immune profile were associated with pCR. A low level of SII+NLR served as an independent predictor of pCR, while distinct TLS features were associated with the presence of pCR. Focusing on the immune profile was crucial for optimal management of gastric cancer patients receiving preoperative treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Haojie Li
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Xuefei Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
25
|
Huang C, Deng M, Leng D, Sun B, Zheng P, Zhang XD. MIRS: An AI scoring system for predicting the prognosis and therapy of breast cancer. iScience 2023; 26:108322. [PMID: 38026206 PMCID: PMC10665820 DOI: 10.1016/j.isci.2023.108322] [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: 10/22/2022] [Revised: 09/25/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Tumor-infiltrating immune cells (TIICs) and metastasis are crucial characteristics for tumorigenesis. However, the potential role of their combination in breast cancer (BRCA) remains elusive. Herein, on the basis of quantifying TIICs and tumor metastasis together, we established a precise prognostic scoring system named metastatic and immunogenomic risk score (MIRS) using a neural network model. MIRS showed better performance when compared with other published signatures. MIRS stratifies patients into a high risk subtype (MIRShigh) and a low risk subtype (MIRSlow). The MIRShigh patients exhibit significantly lower survival rate compared with MIRSlow patients (P < 0.0001 ), higher response to chemotherapy, but lower response to immunotherapy. Conversely, higher infiltration level of TIICs and significantly prolonged survival (P = 0.029 ) are observed in MIRSlow patients, indicating sensitive response in immunotherapy. This work presents a promising indicator to guide treatment options of the BRCA population and provides a predicted webtool that is almost universally applicable to BRCA patients.
Collapse
Affiliation(s)
- Chen Huang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau SAR 999078, China
- State Key laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau SAR 999078, China
| | - Min Deng
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Dongliang Leng
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Baoqing Sun
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Peiyan Zheng
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaohua Douglas Zhang
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
26
|
Zhang K, Xie X, Zheng SL, Deng YR, Liao D, Yan HC, Kang X, Jiang HP, Guo SQ. Tertiary lymphoid structures in gynecological cancers: prognostic role, methods for evaluating, antitumor immunity, and induction for therapy. Front Oncol 2023; 13:1276907. [PMID: 38023214 PMCID: PMC10667730 DOI: 10.3389/fonc.2023.1276907] [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: 08/13/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Tertiary lymphoid structures (TLSs), referred to as tertiary lymphoid organs and lymphoid tissue neogenesis, are aggregates of immune cells that occur in nonlymphoid tissues. In recent years, it has been found that TLSs within the tumor microenvironment have been associated with local adaptive immune immunity against cancer and favorable prognosis in several human solid tumors, including gynecological cancers. The issue of the prognosis of gynecological cancers, including endometrial, cervical, and ovarian cancer, is an enormous challenge that many clinical doctors and researchers are now facing. Concerning the predictive prognostic role of TLSs, effective evaluation, and quantification of TLSs in human tissues may be used to assist gynecologists in assessing the clinical outcome of gynecological cancer patients. This review summarizes the current knowledge of TLSs in gynecological cancers, mainly focusing on the potential mechanism of TLS neogenesis, methods for evaluating TLSs, their prognostic value, and their role in antitumor immune immunity. This review also discusses the new therapeutic methods currently being explored in gynecological cancers to induce the formation of TLSs.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Gynecology, Pingxiang People's Hospital, Pingxiang, Jiangxi, China
| | - Xiao Xie
- Department of Urology, Pingxiang People's Hospital, Pingxiang, Jiangxi, China
| | - Shuang-Lin Zheng
- Department of Gynecology, The Third Hospital of Mianyang, Mianyang, Sichuan, China
| | - Yuan-Run Deng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
- The Third Clinical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Dan Liao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
- The Third Clinical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Hai-Chen Yan
- Department of Urology, Pingxiang People's Hospital, Pingxiang, Jiangxi, China
| | - Xi Kang
- Department of Urology, Pingxiang People's Hospital, Pingxiang, Jiangxi, China
| | - Hui-Ping Jiang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
- The Third Clinical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Sui-Qun Guo
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
- The Third Clinical College, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
27
|
Bao J, Betzler AC, Hess J, Brunner C. Exploring the dual role of B cells in solid tumors: implications for head and neck squamous cell carcinoma. Front Immunol 2023; 14:1233085. [PMID: 37868967 PMCID: PMC10586314 DOI: 10.3389/fimmu.2023.1233085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/06/2023] [Indexed: 10/24/2023] Open
Abstract
In the tumor milieu of head and neck squamous cell carcinoma (HNSCC), distinct B cell subpopulations are present, which exert either pro- or anti-tumor activities. Multiple factors, including hypoxia, cytokines, interactions with tumor cells, and other immune infiltrating lymphocytes (TILs), alter the equilibrium between the dual roles of B cells leading to cancerogenesis. Certain B cell subsets in the tumor microenvironment (TME) exhibit immunosuppressive function. These cells are known as regulatory B (Breg) cells. Breg cells suppress immune responses by secreting a series of immunosuppressive cytokines, including IL-10, IL-35, TGF-β, granzyme B, and adenosine or dampen effector TILs by intercellular contacts. Multiple Breg phenotypes have been discovered in human and mouse cancer models. However, when compartmentalized within a tertiary lymphoid structure (TLS), B cells predominantly play anti-tumor effects. A mature TLS contains a CD20+ B cell zone with several important types of B cells, including germinal-center like B cells, antibody-secreting plasma cells, and memory B cells. They kill tumor cells via antibody-dependent cytotoxicity and phagocytosis, and local complement activation effects. TLSs are also privileged sites for local T and B cell coordination and activation. Nonetheless, in some cases, TLSs may serve as a niche for hidden tumor cells and indicate a bad prognosis. Thus, TIL-B cells exhibit bidirectional immune-modulatory activity and are responsive to a variety of immunotherapies. In this review, we discuss the functional distinctions between immunosuppressive Breg cells and immunogenic effector B cells that mature within TLSs with the focus on tumors of HNSCC patients. Additionally, we review contemporary immunotherapies that aim to target TIL-B cells. For the development of innovative therapeutic approaches to complement T-cell-based immunotherapy, a full understanding of either effector B cells or Breg cells is necessary.
Collapse
Affiliation(s)
- Jiantong Bao
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
- School of Medicine, Southeast University, Nanjing, China
| | - Annika C. Betzler
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Jochen Hess
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cornelia Brunner
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| |
Collapse
|
28
|
Wang L, Geng H, Liu Y, Liu L, Chen Y, Wu F, Liu Z, Ling S, Wang Y, Zhou L. Hot and cold tumors: Immunological features and the therapeutic strategies. MedComm (Beijing) 2023; 4:e343. [PMID: 37638340 PMCID: PMC10458686 DOI: 10.1002/mco2.343] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
The "hotness" or "coldness" of the tumors are determined by the information of the cancer cells themselves, tumor immune characteristics, tumor microenvironment, and signaling mechanisms, which are key factors affecting cancer patients' clinical efficacy. The switch mechanism of "hotness" and "coldness" and its corresponding pathological characteristics and treatment strategies are the frontier and hot spot of tumor treatment. How to distinguish the "hotness" or "coldness" effectively and clarify the causes, microenvironment state, and characteristics are very important for the tumor response and efficacy treatments. Starting from the concept of hot and cold tumor, this review systematically summarized the molecular characteristics, influencing factors, and therapeutic strategies of "hot and cold tumors," and analyzed the immunophenotypes, the tumor microenvironment, the signaling pathways, and the molecular markers that contribute to "hot and cold tumors" in details. Different therapeutic strategies for "cold and hot tumors" based on clinical efficacy were analyzed with drug targets and proteins for "cold and hot tumors." Furthermore, this review combines the therapeutic strategies of different "hot and cold tumors" with traditional medicine and modern medicine, to provide a basis and guidance for clinical decision-making of cancer treatment.
Collapse
Affiliation(s)
- Lianjie Wang
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Hui Geng
- Department of Internal MedicineShanghai International Medical CenterShanghaiChina
| | - Yujie Liu
- Department of NephrologyShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Lei Liu
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yanhua Chen
- Department of the Tumor Research Center, Academy of Integrative MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Fanchen Wu
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zhiyi Liu
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Shiliang Ling
- Department of Medical OncologyNingbo Hospital of Traditional Chinese Medicine, Zhejiang ProvinceNingboChina
| | - Yan Wang
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Lihong Zhou
- Department of Medical Oncology and Cancer InstituteShuguang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
29
|
Yao Y, Xuan H, Wang J, Gong L, Gao W. Integrative analysis of tertiary lymphoid structures and immune microenvironment in patients with esophageal carcinoma. TUMORI JOURNAL 2023; 109:466-480. [PMID: 37249074 DOI: 10.1177/03008916231176857] [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] [Indexed: 05/31/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most common upper gastrointestinal malignancies worldwide. Tertiary lymphoid structures (TLS) are tumor-infiltrating immune cells aggregates coupled with stromal cells which are similar to secondary lymphoid organs. The objective of this study is to explore the predictive effects of two common genes associated with TLS models on prognosis and immunotherapy effects in ESCC patients. METHODS Clinical information for ESCC patients in the TCGA(The Cancer Genome Altas) cohort and GSE 53625 were collected. All of the samples were classified as either high score group or low score group based on two TLS signatures, and the association between TLS signatures and survival, clinical indicators, genomic burden, stemness indices analysis, tumor microenvironment and immunotherapy response were performed. Furthermore, the mature TLS was also assessed in ESCC tissue microarray. RESULTS In our study, we quantified the score of TLS_9 and TLS_12, respectively, reflecting the different statuses of TLS (TLS_9 = B and T cells in TLSs; TLS_12 = neogenesis of TLSs). Subsequently, we explored the effect of TLS score on ESCC tumor microenvironment quantified by multiple algorithms. We found that a correlation analysis indicated that TLS_9 and TLS_12 were all positively correlated with CD8+ T cell, NK cells, CD4+ T cells, M1 macrophages and so on. Meanwhile, some cells present a different correlation pattern of TLS_9 and TLS_12, including activated CD4+ memory T cells and Tgd cells. Immune-related analysis revealed that the TLS_12 and TLS_9 scores were all positively correlated with immune dysfunction, yet negatively correlated with immune exclusion. Following this, the biological roles of TLS_9 and TLS_12 scores were investigated. Also, we noticed that the TLS score could significantly affect the CAFs infiltration and be associated with the genomic burden and tumor stemness. In addition, we explored the prognostic value of mature TLS through tissue microarray (TMA). Our result displayed ESCC patients with the presence of mature TLS had a better prognosis than ESCC patients without it. CONCLUSIONS Our study indicated that ESCC patients with the presence of TLS had better outcomes and an inflamed immune microenvironment. In addition, both TLS-9 and TLS-12 gene signatures could be used as potential biomarkers for the immunotherapy of ESCC patients.
Collapse
Affiliation(s)
- Yuanshan Yao
- Department of Thoracic Surgery, Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Haojie Xuan
- Department of Thoracic Surgery, Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jing Wang
- Department of Thoracic Surgery, Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Libao Gong
- Department of abdominal oncology, The cancer center of the fifth affiliated hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Wen Gao
- Department of Thoracic Surgery, Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
30
|
Yang M, Che Y, Li K, Fang Z, Li S, Wang M, Zhang Y, Xu Z, Luo L, Wu C, Lai X, Wang W. Detection and quantitative analysis of tumor-associated tertiary lymphoid structures. J Zhejiang Univ Sci B 2023; 24:779-795. [PMID: 37701955 PMCID: PMC10500099 DOI: 10.1631/jzus.b2200605] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/27/2023] [Indexed: 09/14/2023]
Abstract
Tumor-associated tertiary lymphoid structures (TLSs) are ectopic lymphoid formations within tumor tissue, with mainly B and T cell populations forming the organic aggregates. The presence of TLSs in tumors has been strongly associated with patient responsiveness to immunotherapy regimens and improving tumor prognosis. Researchers have been motivated to actively explore TLSs due to their bright clinical application prospects. Various studies have attempted to decipher TLSs regarding their formation mechanism, structural composition, induction generation, predictive markers, and clinical utilization. Meanwhile, the scientific approaches to qualitative and quantitative descriptions are crucial for TLS studies. In terms of detection, hematoxylin and eosin (H&E), multiplex immunohistochemistry (mIHC), multiplex immunofluorescence (mIF), and 12-chemokine gene signature have been the top approved methods. However, no standard methods exist for the quantitative analysis of TLSs, such as absolute TLS count, analysis of TLS constituent cells, structural features, TLS spatial location, density, and maturity. This study reviews the latest research progress on TLS detection and quantification, proposes new directions for TLS assessment, and addresses issues for the quantitative application of TLSs in the clinic.
Collapse
Affiliation(s)
- Man Yang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Yurou Che
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Kezhen Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- Department of Oncology, School of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Zengyi Fang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Simin Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- Department of Oncology, School of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Mei Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Yiyao Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Zhu Xu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- Department of Oncology, School of Clinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Liping Luo
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Chuan Wu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Xin Lai
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Weidong Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China.
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China.
- Department of Oncology, School of Clinical Medicine, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
31
|
Zeng L, Koh VCY, Chen XY, Tan PH. Tertiary lymphoid structures in breast ductal carcinoma in situ correlate with adverse pathological parameters. Histopathology 2023; 82:779-788. [PMID: 36635954 DOI: 10.1111/his.14865] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 12/17/2022] [Accepted: 01/07/2023] [Indexed: 01/14/2023]
Abstract
AIMS To investigate tertiary lymphoid structures (TLSs) in ductal carcinoma in situ (DCIS) of the breast and their correlation with pathological features, immune cell markers and clinical outcomes. METHODS AND RESULTS Morphological identification of TLSs in 198 DCIS cases incorporated B and T cell zones with high endothelial venules. TLS positivity was defined as ≥ 1 TLSs in lesional areas, while TLS area percentage was divided into two categories: low (TLSs < 5%) and high (TLSs ≥ 5%). Previously reported biomarkers included ER, PR, HER2, CD68, CD163, CD4, CD8 and PD-L1. TLSs were observed in 24.7% (49 of 198) of cases, with a mean diameter of 0.44 mm (median = 0.4 mm, range = 0.12-1.43 mm). TLSs were significantly associated with higher nuclear grade, presence of necrosis, hormone receptor negativity/HER2 positivity, triple negativity, tumour infiltrating lymphocytes (TILs) and immune related biomarkers such as FOXP3, CD163, CD4 and CD4/CD8 ratio (all P < 0.05). There were no significant associations between TLSs and recurrence, but a combination of TLSshigh with FOXP3+ , CD4high , CD4/CD8 ratiohigh and CD68high individually, compared with all other combinations, disclosed significantly poorer disease-free survival (DFS) for ipsilateral invasive recurrence (IIR) on both Kaplan-Meier and multivariable Cox regression analyses (all P < 0.05). CONCLUSIONS TLSs in DCIS were associated with unfavourable prognostic features, TILs and immune cell markers in our study. TLSshigh /FoxP3+ , TLSshigh /CD4high , TLSshigh /(CD4/CD8) ratiohigh and TLSshigh /CD68high were independent factors for poorer DFS for IIR. Further exploration of the pathological significance of TLSs may provide a clinical basis for their recognition as an important structure and functional unit in the tumour immune microenvironment.
Collapse
Affiliation(s)
- Lixia Zeng
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Department of Pathology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Valerie Cui Yun Koh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Xiao-Yang Chen
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Puay Hoon Tan
- Luma Medical Centre, Singapore, Singapore.,Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore.,Department of Pathology, University of Western Sydney, Sydney, Australia
| |
Collapse
|
32
|
He M, He Q, Cai X, Liu J, Deng H, Li F, Zhong R, Lu Y, Peng H, Wu X, Chen Z, Lao S, Li C, Li J, He J, Liang W. Intratumoral tertiary lymphoid structure (TLS) maturation is influenced by draining lymph nodes of lung cancer. J Immunother Cancer 2023; 11:jitc-2022-005539. [PMID: 37072348 PMCID: PMC10124324 DOI: 10.1136/jitc-2022-005539] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Tertiary lymphoid structure (TLS) is an organized infiltration of immune cells, showing features of germinal center (GC) commonly seen in secondary lymphoid organs. However, its relationship with tumor-draining lymph nodes (TDLNs) has not been studied and we hypothesized that TDLN may influence maturation of intratumoral TLS in non-small cell lung cancer (NSCLC). METHODS Tissue slides of 616 patients that had undergone surgeries were examined. Cox proportional hazard regression model was used to assess risk factors of patients' survival, and logistic regression model was used for their relationship with TLS. Single-cell RNA-sequencing (scRNA-seq) was employed to explore transcriptomic features of TDLNs. Immunohistochemistry, multiplex immunofluorescence and flow cytometry were performed to analyze cellular composition. Cellular components of NSCLC samples from The Cancer Genome Atlas database were inferred with Microenvironment Cell Populations-counter (MCP-counter) method. Murine NSCLC models were used to dissect underlying mechanisms for relationship between TDLN and TLS maturation. RESULTS While GC+ TLS was associated with better prognosis, GC- TLS was not. TDLN metastasis reduced the prognostic relevance of TLS, and was associated with less GC formation. Primary tumor sites showed reduced B cell infiltration in TDLN-positive patients, and scRNA-seq revealed diminished memory B cell formation in tumor-invaded TDLNs, together with an emphasis on weakened interferon (IFN)-γ response. Murine NSCLC models revealed that IFN-γ signaling is involved in memory B cell differentiation in TDLNs and GC formation in primary tumors. CONCLUSIONS Our research emphasizes the influence of TDLN on intratumoral TLS maturation and suggests a role of memory B cells and IFN-γ signaling in this communication.
Collapse
Affiliation(s)
- Miao He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Qihua He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Xiuyu Cai
- Department of General Internal Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jun Liu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Hongshen Deng
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Feng Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Ran Zhong
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Yi Lu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Haoxin Peng
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Xiangrong Wu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Zisheng Chen
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Shen Lao
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Caichen Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Jianfu Li
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
- Department of surgery, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, China
- Department of Medical Oncology, The First People Hospital of Zhaoqing, Zhaoqing, Guangdong, China
| |
Collapse
|
33
|
Zhang D, Wang Y, Zhao F, Yang Q. Integrated multiomics analyses unveil the implication of a costimulatory molecule score on tumor aggressiveness and immune evasion in breast cancer: A large-scale study through over 8,000 patients. Comput Biol Med 2023; 159:106866. [PMID: 37068318 DOI: 10.1016/j.compbiomed.2023.106866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/05/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Although immunotherapy has revolutionised cancer management, reliable genomic biomarkers for identifying eligible patient subpopulations are lacking. Costimulatory molecules play a crucial role in mounting anti-tumour responses, and clinical trials targeting these novel biomarkers are underway. However, whether these molecules can determine tumour aggressiveness and the risk of tumour evasion in breast cancer (BC) remains largely unknown. METHODS The whole-tissue transcriptomic data of 8236 patients with BC from 15 independent cohorts were extracted. An integrated scoring system named 'costimulatory molecule score' (CMS) was constructed and sufficient validated using least absolute shrinkage and selection operator regression (1000 iterations) and the random survival forest algorithm (1000 trees). The correlation among CMSs, cancer genotypes and clinicopathological characteristics was examined. Extensive multiomics and immunogenomic analyses were performed to investigate and verify the association among CMSs, enriched pathways, potential intrinsic and extrinsic immune escape mechanisms, immunotherapy response and therapeutic options. RESULTS The predictive role of CMS model that relies on expression pattern of merely 5 costimulatory genes for prognosis is almost universally applicable to BC patients in a platform-independent manner. Through internal and external in silico validation, high CMS was characterized by favorable genotypes but decreased tumor immunogenicity, activation of stroma, immune-suppressive states and potential immunotherapeutic resistance. Similar results were observed in a real-world immunotherapy cohort and Pan-Cancer analysis. CONCLUSION This comprehensive characterization indicates CMS model may be complemented for predicting tumor aggressiveness and immune evasion in BC patients, underlining the future clinical potential for further exploration of resistance mechanisms and optimization of immunotherapeutic strategies.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China; Department of Clinical Medicine, The First Clinical College, Shandong University, Jinan, 250012, China
| | - Yingnan Wang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China; Department of Clinical Medicine, The First Clinical College, Shandong University, Jinan, 250012, China
| | - Faming Zhao
- Key Laboratory of Environmental Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qifeng Yang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China; Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China; Research Institute of Breast Cancer, Shandong University, Jinan, 250102, China.
| |
Collapse
|
34
|
Falahat R, Berglund A, Perez-Villarroel P, Putney RM, Hamaidi I, Kim S, Pilon-Thomas S, Barber GN, Mulé JJ. Epigenetic state determines the in vivo efficacy of STING agonist therapy. Nat Commun 2023; 14:1573. [PMID: 36949064 PMCID: PMC10033671 DOI: 10.1038/s41467-023-37217-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 03/06/2023] [Indexed: 03/24/2023] Open
Abstract
While STING-activating agents have shown limited efficacy in early-phase clinical trials, multiple lines of evidence suggest the importance of tumor cell-intrinsic STING function in mediating antitumor immune responses. Although STING signaling is impaired in human melanoma, its restoration through epigenetic reprogramming can augment its antigenicity and T cell recognition. In this study, we show that reversal of methylation silencing of STING in murine melanoma cell lines using a clinically available DNA methylation inhibitor can improve agonist-induced STING activation and type-I IFN induction, which, in tumor-bearing mice, can induce tumor regression through a CD8+ T cell-dependent immune response. These findings not only provide mechanistic insight into how STING signaling dysfunction in tumor cells can contribute to impaired responses to STING agonist therapy, but also suggest that pharmacological restoration of STING signaling through epigenetic reprogramming might improve the therapeutic efficacy of STING agonists.
Collapse
Affiliation(s)
- Rana Falahat
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Anders Berglund
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | | | - Ryan M Putney
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Imene Hamaidi
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Sungjune Kim
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, 33612, USA
- Radiation Oncology Program, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Shari Pilon-Thomas
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, 33612, USA
- Cutaneous Oncology Program, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Glen N Barber
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - James J Mulé
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, 33612, USA.
- Radiation Oncology Program, Moffitt Cancer Center, Tampa, FL, 33612, USA.
- Cutaneous Oncology Program, Moffitt Cancer Center, Tampa, FL, 33612, USA.
| |
Collapse
|
35
|
Wang M, Zhai R, Wang M, Zhu W, Zhang J, Yu M, Zhang W, Ye J, Liu L. Tertiary lymphoid structures in head and neck squamous cell carcinoma improve prognosis by recruiting CD8 + T cells. Mol Oncol 2023. [PMID: 36825382 PMCID: PMC10399718 DOI: 10.1002/1878-0261.13403] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 01/21/2023] [Accepted: 02/22/2023] [Indexed: 02/25/2023] Open
Abstract
Tertiary lymphoid structures (TLSs) are formed in long-term chronic inflammation, promoting the local recruitment of lymphocytes, antigen presentation and regulation of immune response, correlated with a better prognosis for cancer patients. Although studies have been conducted to explore methods that accelerate the establishment of TLSs, related research in head and neck squamous cell carcinoma (HNSCC) is still lacking. In this study, we analysed data from The Cancer Genome Atlas and performed immunohistochemical staining analyses of 188 patient samples. The results showed that TLSs promoted the infiltration of immune cells. Patients with TLSs with high infiltration of CD8+ cells showed the best prognosis. Since lymphotoxin α (LTα) was significantly increased in tissues with TLSs, we overexpressed LTα in SCC7 cells (a mouse-derived HNSCC cell line) and established tongue-tumour-bearing models. The polychromatic observation of tissue sections showed that T-cell aggregation increased in the LTα cell group, and a grade 1 TLS was formed on the 12th day after inoculating the cells. Moreover, the tumour volume in the LTα group was significantly less than that of the control group, whereas both the number and the proportion of infiltrated CD8+ T cells were increased. The peripheral CD8+ cells in mice were removed, and no difference was observed in tumour size or TLS formation. Remarkably, we found that TLS led to an increase in the antitumour effect by recruiting CD8+ T cells in HNSCC, showing a CD8+ T-cell-dependent antitumour effect. Moreover, LTα overexpression in the tumour promoted the formation of TLSs.
Collapse
Affiliation(s)
- Mengyao Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, China.,Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, China
| | - Rundong Zhai
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, China.,Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, China
| | - Mengqi Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, China.,Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, China
| | - Weiwen Zhu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, China.,Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, China
| | - Jiayi Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, China.,Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, China
| | - Miao Yu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, China.,Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, China
| | - Wei Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, China.,Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, China
| | - Jinhai Ye
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, China.,Depatment of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, China
| | - Laikui Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, China.,Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, China
| |
Collapse
|
36
|
Tertiary Lymphoid Structures Are Associated with a Favorable Prognosis in High-Grade Serous Ovarian Cancer Patients. Reprod Sci 2023:10.1007/s43032-023-01188-x. [PMID: 36759495 DOI: 10.1007/s43032-023-01188-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
There was accumulating evidence indicating that tertiary lymphoid structures (TLSs) were strongly associated with improved survival and clinical outcome in several solid tumors. In this study, we intended to assess the presence of TLSs and their potential clinical significance in high-grade serous ovarian cancer (HGSOC). TCGA (The Cancer Genome Atlas) cohort included RNA-seq data of 376 HGSOC patients, of which 74 patients included available hematoxylin-eosin (H&E) sections; GEO (Gene Expression Omnibus) cohort, GSE140082, included microarray data of 212 HGSOC patients. TLSs were counted by pathological sections, and the relative abundance of TLSs was assessed by the unsupervised consensus clustering of 12-chemokine transcriptome signatures. The potential associations between TLSs and clinical prognosis, tumor microenvironment (TME), and immunotherapy response of HGSOC were further performed based on transcriptome data. In the H&E sections of HGSOC, TLSs were predominantly located in the stroma and invasive margin of the tumor. Pathological counting results suggested that the expression of 12 chemokines was significantly higher in samples abundant with TLSs than that in the lack of TLSs. Consensus clustering of both TCGA and GEO cohorts divided HGSOC patients into two clusters with different TLSs abundance: low- and high-TLSs. Based on transcriptome analysis, the high-TLS cluster was characterized by better clinical prognosis, a higher degree of immune infiltration, more biological pathways, higher tumor mutational burden score, and higher expression of immune checkpoints. In conclusion, TLSs strongly correlated with the immune-responsive microenvironment and remained a favorable prognostic factor independent of other clinical characteristics in HGSOC. The presence of TLSs was also associated with a potentially favorable response to immune checkpoint blockade (ICB) therapy in HGSOC.
Collapse
|
37
|
Zou X, Liu Y, Lin X, Wang R, Dai Z, Chen Y, Ma M, Tasiheng Y, Yan Y, Wang X, Yu X, Cheng H, Liu C. Characterization of Estrogen Receptors in Pancreatic Adenocarcinoma with Tertiary Lymphoid Structures. Cancers (Basel) 2023; 15:828. [PMID: 36765788 PMCID: PMC9913785 DOI: 10.3390/cancers15030828] [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: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023] Open
Abstract
The role of estrogen signaling in antitumor immunology remains unknown for non-traditional sex-biased cancer types such as pancreatic adenocarcinoma (PAAD). Tertiary lymphoid structures (TLS) are active zones composed of multiple types of immune cells, whose presence indicates anti-tumor immune responses. In this study, we employed a 12-chemokine signature to characterize potential gene categories associated with TLS development and identified seventeen major gene categories including estrogen receptors (ERs). Immunohistochemistry staining revealed the expression patterns of three ERs (ERα, ERβ, and GPER) in 174 PAAD samples, and their correlation with clinicopathological characteristics, immune cell infiltration levels, and intratumoral TLS presence was analyzed. The results indicated that ERα (+) and ERβ (+) were correlated with high tumor grade, and ERβ (+) and GPER (+) were correlated with lower TNM stage, and both ERα (+) and GPER (+) displayed a beneficial effect on prognosis in this cohort. Interestingly, positive staining of all three ERs was significantly correlated with the presence of intratumoral TLSs and infiltration of more active immune cells into the microenvironment. Moreover, the chemotaxis of CD8+T-cells to PAAD cells was significantly increased in vitro with upregulated expression of ERα or ERβ on PAAD cells. To conclude, our study showed a novel correlation between ER expression and TLS development, suggesting that ERs may play a protective role by enhancing anti-tumor immune responses in PAAD.
Collapse
Affiliation(s)
- Xuan Zou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Yu Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Xuan Lin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Ruijie Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Zhengjie Dai
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Yusheng Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Mingjian Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Yesiboli Tasiheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Yu Yan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Xu Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
- Cancer Institute, Shanghai Key Laboratory of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - He Cheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Chen Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| |
Collapse
|
38
|
Liu Z, Meng X, Tang X, Zou W, He Y. Intratumoral tertiary lymphoid structures promote patient survival and immunotherapy response in head neck squamous cell carcinoma. Cancer Immunol Immunother 2022; 72:1505-1521. [DOI: 10.1007/s00262-022-03310-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/11/2022] [Indexed: 12/13/2022]
Abstract
AbstractTertiary lymphoid structures (TLSs) hold the potential role in the prediction of immunotherapy response in several clinical trials. TLSs in head neck squamous cell carcinoma (HNSCC) have been investigated through IHC analysis, whereas there is no TLS gene signature to evaluate the level of TLS neogenesis. We here proposed a TLS signature containing 13 chemokines and determined TLS-hi and TLS-low groups in HNSCC samples from The Cancer Genome Atlas. TLS-hi condition signified a better overall survival. A more inflamed immune infiltrative landscape was identified in the TLS-hi tumors characterized by higher proportion of T cells, TCR/BCR activation and antigen processing. High level of TLSs has a determined role in the clinical significance of T cells. Interesting discovery was that innate lymphoid cells and cancer-associated fibroblasts were positively associated with TLS neogenesis in TME of HNSCC. Furthermore, by integrated TLSs with stromal cells and score, immune cells and score, TMB and malignant cells, we proposed a novel HNSCC TME classifications (HNSCC-TCs 1–5), unravelling the counteracted role of stromal cells and score in inflamed immune landscape, which may provide a novel stromal targeted modality in HNSCC therapy. Finally, we verified that TLS statue is an ideal predictor for immune checkpoint blockade immunotherapy. Current study indicated that the TLSs serve as a novel prognostic biomarker and predictor for immunotherapy, which may provide directions to the current investigations on immunotherapeutic strategies for HNSCC.
Collapse
|
39
|
Tertiary Lymphoid Structures: A Potential Biomarker for Anti-Cancer Therapy. Cancers (Basel) 2022; 14:cancers14235968. [PMID: 36497450 PMCID: PMC9739898 DOI: 10.3390/cancers14235968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
A tertiary lymphoid structure (TLS) is a special component in the immune microenvironment that is mainly composed of tumor-infiltrating lymphocytes (TILs), including T cells, B cells, DC cells, and high endothelial venules (HEVs). For cancer patients, evaluation of the immune microenvironment has a predictive effect on tumor biological behavior, treatment methods, and prognosis. As a result, TLSs have begun to attract the attention of researchers as a new potential biomarker. However, the composition and mechanisms of TLSs are still unclear, and clinical detection methods are still being explored. Although some meaningful results have been obtained in clinical trials, there is still a long way to go before such methods can be applied in clinical practice. However, we believe that with the continuous progress of basic research and clinical trials, TLS detection and related treatment can benefit more and more patients. In this review, we generalize the definition and composition of TLSs, summarize clinical trials involving TLSs according to treatment methods, and describe possible methods of inducing TLS formation.
Collapse
|
40
|
Park S, Cha H, Kim HS, Lee B, Kim S, Kim TM, Jung HA, Sun J, Ahn JS, Ahn M, Park K, Park W, Lee S. Transcriptional upregulation of
CXCL13
is correlated with a favorable response to immune checkpoint inhibitors in lung adenocarcinoma. Cancer Med 2022; 12:7639-7650. [PMID: 36453453 PMCID: PMC10067078 DOI: 10.1002/cam4.5460] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The chemokine CXCL13 is known to influence local anti-tumor immunity by recruiting immune cells and forming tertiary lymphoid structures (TLS). It has been hypothesized that TLS, led by the expression of CXCL13, could be a predictive or prognostic biomarker for immunotherapy. We investigated the predictive value of CXCL13 to immune checkpoint inhibitors (ICI) in lung adenocarcinoma. METHODS We constructed an exploratory dataset (n = 63) and a validation dataset (n = 57) in metastatic lung adenocarcinoma patients treated with ICI. Based on the clinical response, the difference in gene expression profile, including CXCL13, was evaluated. RESULTS From the exploratory dataset, CXCL13 expression was significantly upregulated in the ICI responders (p = 0.002). Survival analysis using a cut-off value of the median expression value of CXCL13 showed prolonged progression-free survival (PFS) (p = 0.004) and overall survival (OS) (p = 0.007). CXCL13 expression was correlated with other immune response genes, such as GZMA, CD8A, IFNG, PRF1, TLS-related gene sets and its receptor, CXCR5. Notably, subgroup analyses based on CXCL13 expression and CD8A showed that CXCL13-upregulated patients demonstrated comparably prolonged survival regardless of CD8A expression. In the validation dataset, CXCL13 upregulation also demonstrated a significant prolongation of both PFS (p = 0.050) and OS (p = 0.026). CONCLUSION We observed that CXCL13 upregulation is correlated to better ICI response in lung adenocarcinoma. Our results support that CXCL13 could be an important chemokine in shaping the immunoactive tumor microenvironment which affects the anti-tumor effect of ICI.
Collapse
Affiliation(s)
- Sehhoon Park
- Division of Hematology‐Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Hongui Cha
- Department of Health Science and Technology, Samsung Advanced Institute of Health Science and Technology Sungkyunkwan University Seoul Republic of Korea
- Samsung Genome Institute Samsung Medical Center, Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Hong Sook Kim
- Department of Health Science and Technology, Samsung Advanced Institute of Health Science and Technology Sungkyunkwan University Seoul Republic of Korea
| | - Boram Lee
- Department of Health Science and Technology, Samsung Advanced Institute of Health Science and Technology Sungkyunkwan University Seoul Republic of Korea
- Samsung Genome Institute Samsung Medical Center, Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Soyeon Kim
- Seoul National University Cancer Research Institute Seoul Republic of Korea
- Biomedical Research Institute, Seoul National University Hospital Seoul Republic of Korea
| | - Tae Min Kim
- Seoul National University Cancer Research Institute Seoul Republic of Korea
- Departments of Internal Medicine Seoul National University Hospital, Seoul National University College of Medicine Seoul Republic of Korea
| | - Hyu Ae Jung
- Division of Hematology‐Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Jong‐Mu Sun
- Division of Hematology‐Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology‐Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Myung‐Ju Ahn
- Division of Hematology‐Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Keunchil Park
- Division of Hematology‐Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine Seoul Republic of Korea
- Department of Health Science and Technology, Samsung Advanced Institute of Health Science and Technology Sungkyunkwan University Seoul Republic of Korea
| | - Woong‐Yang Park
- Department of Health Science and Technology, Samsung Advanced Institute of Health Science and Technology Sungkyunkwan University Seoul Republic of Korea
- Samsung Genome Institute Samsung Medical Center, Sungkyunkwan University School of Medicine Seoul Republic of Korea
| | - Se‐Hoon Lee
- Division of Hematology‐Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine Seoul Republic of Korea
- Department of Health Science and Technology, Samsung Advanced Institute of Health Science and Technology Sungkyunkwan University Seoul Republic of Korea
| |
Collapse
|
41
|
Gou Q, Liu Z, Xie Y, Deng Y, Ma J, Li J, Zheng H. Systematic evaluation of tumor microenvironment and construction of a machine learning model to predict prognosis and immunotherapy efficacy in triple-negative breast cancer based on data mining and sequencing validation. Front Pharmacol 2022; 13:995555. [PMID: 36225561 PMCID: PMC9548553 DOI: 10.3389/fphar.2022.995555] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The role of the tumor microenvironment (TME) in predicting prognosis and therapeutic efficacy has been demonstrated. Nonetheless, no systematic studies have focused on TME patterns or their function in the effectiveness of immunotherapy in triple-negative breast cancer. Methods: We comprehensively estimated the TME infiltration patterns of 491 TNBC patients from four independent cohorts, and three cohorts that received immunotherapy were used for validation. The TME subtypes were comprehensively evaluated based on immune cell infiltration levels in TNBC, and the TRG score was identified and systematically correlated with representative tumor characteristics. We sequenced 80 TNBC samples as an external validation cohort to make our conclusions more convincing. Results: Two TME subtypes were identified and were highly correlated with immune cell infiltration levels and immune-related pathways. More representative TME-related gene (TRG) scores calculated by machine learning could reflect the fundamental characteristics of TME subtypes and predict the efficacy of immunotherapy and the prognosis of TNBC patients. A low TRG score, characterized by activation of immunity and ferroptosis, indicated an activated TME phenotype and better prognosis. A low TRG score showed a better response to immunotherapy in TNBC by TIDE (Tumor Immune Dysfunction and Exclusion) analysis and sensitivity to multiple drugs in GDSC (Genomics of Drug Sensitivity in Cancer) analysis and a significant therapeutic advantage in patients in the three immunotherapy cohorts. Conclusion: TME subtypes played an essential role in assessing the diversity and complexity of the TME in TNBC. The TRG score could be used to evaluate the TME of an individual tumor to enhance our understanding of the TME and guide more effective immunotherapy strategies.
Collapse
Affiliation(s)
- Qiheng Gou
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zijian Liu
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxin Xie
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yuxin Xie,
| | - Yulan Deng
- Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Ji Ma
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiangping Li
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zheng
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
42
|
Prognostic value, DNA variation and immunologic features of a tertiary lymphoid structure-related chemokine signature in clear cell renal cell carcinoma. Cancer Immunol Immunother 2022; 71:1923-1935. [PMID: 35043231 DOI: 10.1007/s00262-021-03123-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/24/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND The tumor microenvironment (TME) and tertiary lymphoid structures (TLS) affect the occurrence and development of cancers. How the immune contexture interacts with the phenotype of clear cell renal cell carcinoma (ccRCC) remains unclear. METHODS We identified and evaluated TLS clusters in ccRCC using machine learning algorithms and the 12-chemokine gene signature for TLS. Analyses for functional enrichment, DNA variation, immune cell distribution, association with independent clinicopathological features and predictive value of CXCL13 in ccRCC were performed. RESULTS We found a prominently enrichment of the 12-chemokine gene signature for TLS in patients with ccRCC compared with other types of renal cell carcinoma. We identified a prognostic value of CCL4, CCL5, CCL8, CCL19 and CXCL13 expression in ccRCC. DNA deletion of the TLS gene signature significantly predicted poor outcome in ccRCC compared with amplification and wild-type gene signature. We established TLS clusters (C1-4) and observed distinct differences in survival, stem cell-like characteristics, immune cell distribution, response to immunotherapies and VEGF-targeted therapies among the clusters. We found that elevated CXCL13 expression significantly predicted aggressive progression and poor prognosis in 232 patients with ccRCC in a real-world validation cohort. CONCLUSION This study described a 12-chemokine gene signature for TLS in ccRCC and established TLS clusters that reflected different TME immune status and corresponded to prognosis of ccRCC. We confirmed the dense presence of TILs aggregation and TLS in ccRCC and demonstrated an oncogenic role of CXCL13 expression of ccRCC, which help develop immunotherapies and provide novel insights on the long-term management of ccRCC.
Collapse
|
43
|
Moragon S, Hernando C, Martinez-Martinez MT, Tapia M, Ortega-Morillo B, Lluch A, Bermejo B, Cejalvo JM. Immunological Landscape of HER-2 Positive Breast Cancer. Cancers (Basel) 2022; 14:3167. [PMID: 35804943 PMCID: PMC9265068 DOI: 10.3390/cancers14133167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
Understanding the biological aspects of immune response in HER2+ breast cancer is crucial to implementing new treatment strategies in these patients. It is well known that anti-HER2 therapy has improved survival in this population, yet a substantial percentage may relapse, creating a need within the scientific community to uncover resistance mechanisms and determine how to overcome them. This systematic review indicates the immunological mechanisms through which trastuzumab and other agents target cancer cells, also outlining the main trials studying immune checkpoint blockade. Finally, we report on anti-HER2 vaccines and include a figure exemplifying their mechanisms of action.
Collapse
Affiliation(s)
- Santiago Moragon
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
| | - Cristina Hernando
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
| | - Maria Teresa Martinez-Martinez
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
| | - Marta Tapia
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
| | - Belen Ortega-Morillo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
| | - Ana Lluch
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
- Instituto de Salud Carlos III, CIBERONC (Centro De Investigacion Biomedica En Red De Cancer), 28220 Madrid, Spain
| | - Begoña Bermejo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
- Instituto de Salud Carlos III, CIBERONC (Centro De Investigacion Biomedica En Red De Cancer), 28220 Madrid, Spain
| | - Juan Miguel Cejalvo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain; (S.M.); (C.H.); (M.T.M.-M.); (M.T.); (B.O.-M.); (A.L.); (B.B.)
- Instituto de Salud Carlos III, CIBERONC (Centro De Investigacion Biomedica En Red De Cancer), 28220 Madrid, Spain
| |
Collapse
|
44
|
Tooley KA, Escobar G, Anderson AC. Spatial determinants of CD8+ T cell differentiation in cancer. Trends Cancer 2022; 8:642-654. [PMID: 35527216 PMCID: PMC9308689 DOI: 10.1016/j.trecan.2022.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 12/25/2022]
Abstract
Uncovering the mechanisms that control CD8+ T cell function is a major focus of cancer research. Advances in flow cytometry and single-cell transcriptomics have provided unprecedented in-depth resolution of CD8+ T cell states in cancer. However, these technologies fail to capture important spatial information, including cell-cell interactions and tissue localization. The discovery that intra-tumoral immune niches, tertiary lymphoid structures, and the tumor-draining lymph node are key sites of inter-cellular communication has evoked interest in understanding the spatial determinants that regulate CD8+ T cell functions at these sites. We focus on the cellular, as well as the soluble and surface-bound signals that regulate CD8+ T cell phenotypes and functions in a topologically-regulated manner, highlighting where new spatial transcriptomics and imaging technologies can uncover mechanistic insights.
Collapse
|
45
|
Spatial charting of single-cell transcriptomes in tissues. Nat Biotechnol 2022; 40:1190-1199. [PMID: 35314812 PMCID: PMC9673606 DOI: 10.1038/s41587-022-01233-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022]
Abstract
Single-cell RNA sequencing methods can profile the transcriptomes of single cells but cannot preserve spatial information. Conversely, spatial transcriptomics assays can profile spatial regions in tissue sections, but do not have single-cell resolution. Here, we developed a computational method called CellTrek that combines these two datasets to achieve single-cell spatial mapping through coembedding and metric learning approaches. We benchmarked CellTrek using simulation and in situ hybridization datasets, which demonstrated its accuracy and robustness. We then applied CellTrek to existing mouse brain and kidney datasets and showed that CellTrek can detect topological patterns of different cell types and cell states. We performed single-cell RNA sequencing and spatial transcriptomics experiments on two ductal carcinoma in situ tissues and applied CellTrek to identify tumor subclones that were restricted to different ducts, and specific T cell states adjacent to the tumor areas. Our data show that CellTrek can accurately map single cells in diverse tissue types to resolve their spatial organization.
Collapse
|
46
|
A 12-chemokine gene signature is associated with the enhanced immunogram scores and is relevant for precision immunotherapy. Med Oncol 2022; 39:43. [PMID: 35092511 DOI: 10.1007/s12032-021-01635-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/21/2021] [Indexed: 10/19/2022]
Abstract
Advances in the understanding of checkpoint blockade immunotherapy have suggested that boosting the cancer-immunity cycle (CIC) can help induce regression of tumors. However, good efficacy only occurs in a subset of patients. Predictive biomarkers that can reflect the tumor microenvironment (TME) and CIC may have great potential. More recently, the presence of intratumoral tertiary lymphoid structures (TLSs) has also been correlated with clinical benefit in patients. In this study, we comprehensively measured the immunogram scores (IGSs) for the CIC and explored the associations between immunological and mutational features and a 12-chemokine metagene TLS signature in data from The Cancer Genome Atlas (TCGA). Three immunotherapy datasets were further applied for validation. In the TCGA dataset, we observed that the 12-chemokine TLS signature score was positively associated with the enhanced IGSs as represented by increased tumor mutational burden (TMB) and neoantigen burden (TNB), enriched immune cell (IC) infiltration, and elevated cytolytic activity and checkpoint expression. Specifically, in bladder cancer and melanoma, the high 12-chemokine TLS signature score was found to potentially reflect an expanded cancer-immunity status characterized by high TNB and an immune-inflamed feature. The predictive and prognostic value of the 12-chemokine TLS signature was further validated in several immunotherapy datasets. The score of the 12-chemokine TLS signature may serve as a pancancer marker of the immune-active phenotype. The 12-chemokine TLS signature showed promise as a predictive and prognostic biomarker for ICB efficacy, especially in melanoma and bladder cancer.
Collapse
|
47
|
Abstract
Ectopic lymphoid aggregates, termed tertiary lymphoid structures (TLSs), are formed in numerous cancer types, and, with few exceptions, their presence is associated with superior prognosis and response to immunotherapy. In spite of their presumed importance, the triggers that lead to TLS formation in cancer tissue and the contribution of these structures to intratumoral immune responses remain incompletely understood. Here, we discuss the present knowledge on TLSs in cancer, focusing on (i) the drivers of TLS formation, (ii) the function and contribution of TLSs to the antitumor immune response, and (iii) the potential of TLSs as therapeutic targets in human cancers.
Collapse
Affiliation(s)
- Ton N Schumacher
- Division of Molecular Oncology and Immunology, Oncode Institute, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Daniela S Thommen
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| |
Collapse
|
48
|
陈 红, 胡 翔, 张 惠, 胡 洪. [Induction and Anti-Tumor Function of Tertiary Lymphoid Organs]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2022; 53:35-42. [PMID: 35048597 PMCID: PMC10408855 DOI: 10.12182/20220160102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To induce the development of tertiary lymphoid organs (TLO) in a mouse model of melanoma and to evaluate TLO's functions in antitumor immunity. METHODS Lymphotoxin-beta receptor (LTβR) was overexpressed in NIH3T3 cells through the lentivirus system and the overexpression efficiency of LTβR in LTβR-NIH3T3 cells was examined. Western blot and qPCR were used to examine the non-canonical nuclear factor (NF)-κB signaling pathway in NIH3T3 cells overexpressing LTβR. B16-OVA melanoma mouse model was constructed to explore the induction of TLO and anti-tumor functions of TLO in LTβR-NIH3T3 cells. RESULTS LTβR was overexpressed in NIH3T3 cells through the lentivirus system, and flow cytometry showed that the proportion of GFP + cells reached 99%. The overexpression of LTβR activated the non-canonical NF-κB signaling pathway in NIH3T3 cells. Findings from the mouse tumor model suggest that the injection of LTβR-NIH3T3 cells successfully induced the development of lymphoid tissue around the tumor and enhanced the tumor infiltration of T cells and MHCⅡ + macrophages, significantly inhibiting tumor growth and prolonging the survival of tumor-bearing mice. CONCLUSION LTβR-NIH3T3 cells promoted anti-tumor immunity by inducing TLO development, which may provide new perspectives for tumor immunotherapy.
Collapse
Affiliation(s)
- 红 陈
- 四川大学华西医院 生物治疗国家重点实验室 (成都 610041)State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- 四川大学华西医院 免疫和血液研究中心 (成都 610041)Center for Immunology and Hematology Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 翔 胡
- 四川大学华西医院 生物治疗国家重点实验室 (成都 610041)State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 惠媛 张
- 四川大学华西医院 生物治疗国家重点实验室 (成都 610041)State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- 四川大学华西医院 免疫和血液研究中心 (成都 610041)Center for Immunology and Hematology Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 洪波 胡
- 四川大学华西医院 生物治疗国家重点实验室 (成都 610041)State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- 四川大学华西医院 免疫和血液研究中心 (成都 610041)Center for Immunology and Hematology Research, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
49
|
Gambardella V, Martínez-Ciarpaglini C, Fleitas T, Cervantes A. In the literature: October 2021. ESMO Open 2021; 6:100285. [PMID: 34752996 PMCID: PMC8586749 DOI: 10.1016/j.esmoop.2021.100285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 11/21/2022] Open
Affiliation(s)
- V Gambardella
- Department of Medical Oncology, Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - C Martínez-Ciarpaglini
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain; Department of Pathology, Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - T Fleitas
- Department of Medical Oncology, Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - A Cervantes
- Department of Medical Oncology, Hospital Clínico Universitario, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
50
|
Gavrielatou N, Vathiotis I, Economopoulou P, Psyrri A. The Role of B Cells in Head and Neck Cancer. Cancers (Basel) 2021; 13:cancers13215383. [PMID: 34771546 PMCID: PMC8582491 DOI: 10.3390/cancers13215383] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 01/13/2023] Open
Abstract
Simple Summary Host immunity has established its role in deciding the course of cancer evolution. As cellular and molecular components in the tumor microenvironment peripherally appear to be at a constant interplay, favoring either tumor control or progression, it is vital to decrypt the immunity elements, which demonstrate the potential to be harnessed towards cancer elimination. Head and neck cancer has been characterized as densely immune infiltrated but at the same time a highly immunosuppressive malignancy due to a negative equilibrium between active and dysfunctional immune cell populations. B-cells constitute the cornerstone of humoral immunity; however, their role in cancer has been vastly overlooked in comparison to other immune subtypes and reports from multiple studies fail to show agreement on their prognostic impact. This review focuses on the role of B-cells on head and neck cancer with the aim to highlight their effect on anti-cancer immunity, as well as their possible impact on immunotherapy outcomes. Abstract Head and neck cancer comprises a heterogenous, highly immune infiltrated malignancy, defined by a predominantly immunosuppressive tumor microenvironment (TME). In recent years, PD-1/PD-L1 immune checkpoint inhibitors have become the standard of care treatment, either as monotherapy or in combination with chemotherapy agents, thus revolutionizing the therapeutic landscape of recurrent/metastatic disease. As a result, preclinical research is increasingly focusing on TME composition and pathophysiology, aiming to comprehensively characterize the specific elements and interactions affecting anti-tumor immunity, as well as to unveil novel predictive biomarkers of immunotherapy outcomes. While T lymphocytic populations have been vastly explored regarding their effect on cancer development, B-cells constitute a far less investigated, yet possibly equally important, aspect of cancer immunity. B-cell presence, either as single cells or as part of tertiary lymphoid structures within the TME, has been associated with several anti-tumor defense mechanisms, such as antigen presentation, antibody production and participation in antibody-dependent cellular cytotoxicity, and has demonstrated prognostic significance for multiple types of malignancies. However, immunoregulatory B-cell phenotypes have also been identified both peripherally and within malignant tissue, bearing inhibitory effects on numerous immune response processes. Consequently, B-cells and their subsets demonstrate the potential to become valuable cancer biomarkers and acquire a leading role in future therapeutic strategies.
Collapse
Affiliation(s)
- Niki Gavrielatou
- Department of Pathology, School of Medicine, Yale University, New Haven, CT 06510, USA; (N.G.); (I.V.)
| | - Ioannis Vathiotis
- Department of Pathology, School of Medicine, Yale University, New Haven, CT 06510, USA; (N.G.); (I.V.)
| | - Panagiota Economopoulou
- Section of Medical Oncology, Second Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Amanda Psyrri
- Section of Medical Oncology, Second Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
- Correspondence:
| |
Collapse
|