1
|
Gao D. The role of non-malignant B cells in malignant hematologic diseases. Hematology 2025; 30:2466261. [PMID: 39964954 DOI: 10.1080/16078454.2025.2466261] [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: 06/02/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
The tumor microenvironment (TME) represents a heterogeneous, complicated ecosystem characterized by intricate interactions between tumor cells and immune cells. During the past decade, immune cells especially T cells were found to play an important role in the progression of tumor and many related immune checkpoints drugs were created. In recent years, more and more scientists revealed the critical role of B-cells within the TME, particularly various populations of non-malignant B cells. Some studies indicated that non-malignant B cells may exert a 'double-edged sword' role in solid tumors. However, there has been comparatively less focus on the role of non-malignant B cells in hematologic malignancies. In this review, we characterized the development of B cells and summarized its functions of antitumor immunity within TME, with an emphasis on elucidating the roles and potential mechanisms of non-malignant B cells in the progression of hematologic diseases including classical Hodgkin's lymphoma, non-Hodgkin's B-cell lymphoma, non-Hodgkin's T-cell lymphoma, leukemia and multiple myeloma.
Collapse
Affiliation(s)
- Daquan Gao
- Department of Hematology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, People's Republic of China
| |
Collapse
|
2
|
Laurent PA, André F, Bobard A, Deandreis D, Demaria S, Depil S, Eichmüller SB, Fernandez-Palomo C, Foijer F, Galluzzi L, Galon J, Guckenberger M, Harrington KJ, Herrera FG, Huber PE, Italiano A, Karam SD, Kroemer G, Lambin P, Leuschner C, Mantovani A, Meylan E, Mondini M, Pittet MJ, Pouget JP, Remon J, Sørensen CS, Sotiriou C, Vanpouille-Box C, Weichselbaum RR, Welsh JW, Zitvogel L, Formenti SC, Deutsch E. Pushing the boundaries of radiotherapy-immunotherapy combinations: highlights from the 7 th immunorad conference. Oncoimmunology 2025; 14:2432726. [PMID: 39696783 DOI: 10.1080/2162402x.2024.2432726] [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: 07/25/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Over the last decade, the annual Immunorad Conference, held under the joint auspicies of Gustave Roussy (Villejuif, France) and the Weill Cornell Medical College (New-York, USA) has aimed at exploring the latest advancements in the fields of tumor immunology and radiotherapy-immunotherapy combinations for the treatment of cancer. Gathering medical oncologists, radiation oncologists, physicians and researchers with esteemed expertise in these fields, the Immunorad Conference bridges the gap between preclinical outcomes and clinical opportunities. Thus, it paves a promising way toward optimizing radiotherapy-immunotherapy combinations and, from a broader perspective, improving therapeutic strategies for patients with cancer. Herein, we report on the topics developed by key-opinion leaders during the 7th Immunorad Conference held in Paris-Les Cordeliers (France) from September 27th to 29th 2023, and set the stage for the 8th edition of Immunorad which will be held at Weill Cornell Medical College (New-York, USA) in October 2024.
Collapse
Affiliation(s)
- Pierre-Antoine Laurent
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- INSERM, U1030 "Molecular Radiotherapy and Therapeutic Innovations", Gustave Roussy, Villejuif, France
| | - Fabrice André
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
- INSERM U981 "Molecular predictors and new targets in oncology", Gustave Roussy, Villejuif, France
- IHU PRISM Precision Medicine Cancer Center, Gustave Roussy, Villejuif, France
| | | | | | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New-York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New-York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Stephane Depil
- Cancer Research Center of Lyon, Centre Léon Bérard, Université Claude Bernard, Lyon, France
- ErVimmune, Lyon, France
| | - Stefan B Eichmüller
- Research Group GMP & T-cell therapy, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | | | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medicine, New-York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology; Sorbonne Université; Sorbonne Paris Cité, Université de Paris, Paris, France
- Centre de Recherche des Cordeliers, Paris, France
| | | | - Kevin J Harrington
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, National Institute of Health Research Biomedical Research Centre, London, UK
| | - Fernanda G Herrera
- Radiation Oncology Service, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
- Immuno-oncology Service, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Peter E Huber
- Department of Radio-oncology and Radiotherapy, University Hospital Heidelberg; Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Antoine Italiano
- Department of therapeutic innovations (DITEP), Gustave Roussy, Villejuif, France
- Department of Medicine, Institut Bergonié, Bordeaux, France
- Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Department of Biology, Hôpital Européen Georges Pompidou AP-HP, Paris, France
- Institut du Cancer Paris CARPEM, Paris, France
| | - Philippe Lambin
- Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- Department of Radiology and Nuclear Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Carola Leuschner
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alberto Mantovani
- IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
- William Harvey Research Institute, Queen Mary University, London, UK
| | - Etienne Meylan
- Laboratory of Immunobiology, Department of Molecular Biology, Faculty of Sciences, Université Libre de Bruxelles, Bruxelles, Belgium
- Lung Cancer and Immuno-Oncology laboratory, Bordet Cancer Research Laboratories, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Faculty of Medicine, Université libre de Bruxelles, Bruxelles, Belgium
- ULB Cancer Research Center (U-CRC) and ULB Center for Research in Immunology (U-CRI), Bruxelles, Belgium
| | - Michele Mondini
- INSERM, U1030 "Molecular Radiotherapy and Therapeutic Innovations", Gustave Roussy, Villejuif, France
| | - Mikael J Pittet
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland. Swiss Cancer Center Leman, Lausanne, Switzerland
- Translational Research Center in Onco-Haematology (CRTOH), University of Geneva, Geneva, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Jean-Pierre Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM)INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Jordi Remon
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Claus S Sørensen
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Claire Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medicine, New-York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, Ludwig Center for Metastasis Research; University of Chicago, Chicago, IL, USA
| | - James W Welsh
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laurence Zitvogel
- ClinicObiome, Gustave Roussy, Villejuif, France
- INSERM U1015 "Tumor Immunology and Anti-Cancer Immunotherapy Unit", Gustave Roussy, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France
- Division of Medicine, Paris-Saclay University, Ile-de-France, France
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New-York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- INSERM, U1030 "Molecular Radiotherapy and Therapeutic Innovations", Gustave Roussy, Villejuif, France
- Division of Medicine, Paris-Saclay University, Ile-de-France, France
- RHU LySAIRI "Lymphocyte-Sparing Artificial Intelligence-guided Radio-Immunotherapy", Gustave Roussy, Villejuif, France
| |
Collapse
|
3
|
Ruiz-Torres DA, Bryan ME, Hirayama S, Merkin RD, Luciani E, Roberts TJ, Patel M, Park JC, Wirth LJ, Sadow PM, Sade-Feldman M, Stott SL, Faden DL. Spatial characterization of tertiary lymphoid structures as predictive biomarkers for immune checkpoint blockade in head and neck squamous cell carcinoma. Oncoimmunology 2025; 14:2466308. [PMID: 39963988 PMCID: PMC11845054 DOI: 10.1080/2162402x.2025.2466308] [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: 11/13/2024] [Revised: 01/27/2025] [Accepted: 02/08/2025] [Indexed: 02/23/2025] Open
Abstract
Immune checkpoint blockade (ICB) is the standard of care for recurrent/metastatic head and neck squamous cell carcinoma (HNSCC), yet efficacy remains low. The combined positive score (CPS) for PD-L1 is the only biomarker approved to predict response to ICB and has limited performance. Tertiary Lymphoid Structures (TLS) have shown promising potential for predicting response to ICB. However, their exact composition, size, and spatial biology in HNSCC remain understudied. To elucidate the impact of TLS spatial biology in response to ICB, we utilized pre-ICB tumor tissue sections from 9 responders (complete response, partial response, or stable disease) and 11 non-responders (progressive disease) classified via RECISTv1.1. A custom multi-immunofluorescence (mIF) staining assay was applied to characterize tumor cells (pan-cytokeratin), T cells (CD4, CD8), B cells (CD19, CD20), myeloid cells (CD16, CD56, CD163), dendritic cells (LAMP3), fibroblasts (α Smooth Muscle Actin), proliferative status (Ki67) and immunoregulatory molecules (PD1). A machine learning model was employed to measure the effect of spatial metrics on achieving a response to ICB. A higher density of B cells (CD20+) was found in responders compared to non-responders to ICB (p = 0.022). The presence of TLS within 100 µm of the tumor was associated with improved overall (p = 0.04) and progression-free survival (p = 0.03). A multivariate machine learning model identified TLS density as a leading predictor of response to ICB with 80% accuracy. Immune cell densities and TLS spatial location play a critical role in the response to ICB in HNSCC and may potentially outperform CPS as a predictor of response.
Collapse
Affiliation(s)
- Daniel A. Ruiz-Torres
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael E. Bryan
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shun Hirayama
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
| | - Ross D. Merkin
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Evelyn Luciani
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Thomas J. Roberts
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Manisha Patel
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Jong C. Park
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Lori J. Wirth
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Peter M. Sadow
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Moshe Sade-Feldman
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shannon L. Stott
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Engineering in Medicine and BioMEMS Resource Center, Surgical Services, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Daniel L. Faden
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| |
Collapse
|
4
|
Chen Z, Tai Y, Deng C, Sun Y, Chen H, Luo T, Lin J, Chen W, Xu H, Song G, Tang Q, Lu J, Zhu X, Wen S, Wang J. Innovative sarcoma therapy using multifaceted nano-PROTAC-induced EZH2 degradation and immunity enhancement. Biomaterials 2025; 321:123344. [PMID: 40262462 DOI: 10.1016/j.biomaterials.2025.123344] [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: 11/03/2024] [Revised: 04/05/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Sarcomas are highly malignant tumors characterized by their heterogeneity and resistance to conventional therapies, which significantly limit treatment options. EZH2 is highly expressed in sarcomas, but targeting it is difficult. In this study, we uncovered the non-canonical transcriptional mechanisms of EZH2 in sarcoma and highlighted the essential role of EZH2 in regulating YAP1 through non-canonical transcriptional pathways in the progression of sarcoma. Building on this, we developed YM@VBM, a novel and versatile nano-PROTAC (proteolysis-targeting chimera), by integrating a polyphenol-vanadium oxide system with the EZH2 degrader YM281 PROTAC, encapsulated in methoxy polyethylene glycol-NH2 to enhance biocompatibility. To further facilitate targeted drug delivery to tumors, YM@VBM nano-PROTACs were incorporated into microneedle patches. Our engineered YM@VBM exhibited multiple functionalities, including the peroxidase-like activity to generate reactive oxygen species, depletion of glutathione, and photothermal effects, specifically targeting sarcoma characteristics. YM@VBM significantly enhanced targeting efficacy via inducing potent EZH2 degradation. Most importantly, it can also activate anti-tumor immunity via excluding myeloid-derived suppressor cells, maturing dendritic cells, and forming tertiary lymphoid structures. Hence, we reveal that YM@VBM presents a promising treatment strategy for sarcoma, offering a multifaceted approach to combat this challenging malignancy.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Yi Tai
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China; Surgical Department of Colorectal Cancer, Zhejiang Cancer Hospital, 1st BanShan East Road, Gongshu District, Hangzhou, 310000, Zhejiang Province, PR China
| | - Chuangzhong Deng
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Yameng Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Hongmin Chen
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Tianqi Luo
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Jiaming Lin
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Weiqing Chen
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Huaiyuan Xu
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Guohui Song
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Qinglian Tang
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Jinchang Lu
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Xiaojun Zhu
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China.
| | - Jin Wang
- Department of Musculoskeletal Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, PR China.
| |
Collapse
|
5
|
Cui M, Zhou M, Zhou L, Zhou G, Liu Y. Tertiary lymphoid structures achieve 'cold' to 'hot' transition by remodeling the cold tumor microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189312. [PMID: 40189114 DOI: 10.1016/j.bbcan.2025.189312] [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: 06/11/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025]
Abstract
Immune checkpoint blockade (ICB) therapies have demonstrated significant clinical efficacy in immune-infiltrated tumors such as melanoma and non-small cell lung cancer. However, "cold tumors"-including ovarian cancer, pancreatic cancer, and gliomas-exhibit insufficient immune infiltration, leading to poor therapeutic responses to ICBs and limited improvement in patient prognosis. Recent studies have shown that tumor-associated tertiary lymphoid structures (TLSs) can induce strong local immune responses within the tumor microenvironment (TME), serving as important biological markers for predicting ICB therapy efficacy. Notably, preclinical and clinical studies on cold tumors have confirmed that TLSs can potently enhance ICB efficacy through TME remodeling-a breakthrough that has attracted considerable attention. Here, we systematically examine the immunological profile of cold tumors and decipher the mechanistic basis for their impaired immune cell infiltration. We further delineate the distinctive features of tumor-associated TLSs in generating antitumor immunity and establish criteria for their identification. Significantly, we emphasize the unique capability of TLSs to reprogram the immunosuppressive tumor microenvironment characteristic of cold tumors. Based on these insights, we evaluate clinical evidence supporting TLS-mediated enhancement of ICB efficacy and discuss emerging strategies for exogenous TLSs induction.
Collapse
Affiliation(s)
- Mengke Cui
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Mengfan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Lu Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Gan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China; National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, 110 Xiangya Road, Changsha, Hunan 410008, PR China.
| | - Yingzi Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China.
| |
Collapse
|
6
|
Li Y, Ma K, Wang H, Liu Z, Li Z. Identification of therapeutic targets in lung adenocarcinoma using Mendelian randomization and multi-omics. Discov Oncol 2025; 16:1028. [PMID: 40481979 PMCID: PMC12145347 DOI: 10.1007/s12672-025-02835-2] [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: 02/27/2025] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) remains associated with limited effective pharmacological treatment options. This study aimed to identify potential therapeutic targets for LUAD through the integration and analysis of multi-omics datasets. METHODS A meta-analysis was conducted using two extensive proteomics datasets, the UK Biobank Proteomics Project (UKB-PPP) and the Fenland study, to identify disease-associated targets for LUAD through the Summary-Data-Based Mendelian Randomization method. Sensitivity analysis, including heterogeneity tests for dependent instruments, were conducted to validate the findings. The prognostic relevance of the identified candidate targets was assessed using transcriptomic data. Functional interactions were explored via protein-protein interaction network analysis, while single-cell analyses were employed to determine cell-specific expression patterns and differentiation trajectories. Potential side effects and therapeutic indications of these targets were evaluated using phenome-wide association studies and pharmacological data mining. RESULTS Following meta-analysis, a primary significant target, intercellular adhesion molecule 5 (ICAM5), along with potential targets FUT8 and KLK13, were identified as therapeutic candidates for LUAD. FUT8 demonstrated a positive association with LUAD risk (OR = 1.02, p = 0.049), while ICAM5 (OR = 0.88, p = 0.002) and KLK13 (OR = 0.85, p = 0.021) exhibited negative associations. ICAM5 was further identified as an independent prognostic factor for patient survival (HR: 0.788, 95% CI: 0.663-0.936, p = 0.007) and revealed significant diagnostic and prognostic utility in LUAD. ICAM5 expression correlated with various immune infiltration patterns, suggesting potential modulation of the tumor immune microenvironment. Single-cell analysis revealed that ICAM5 did not directly impact LUAD cell differentiation, though its downstream target, MUC1, may contribute to differentiation processes, particularly in KRAS-mutated LUAD. Furthermore, phenome-wide association studies did not reveal substantial evidence of adverse phenotypes linked to ICAM5, supporting its safety profile for drug development. CONCLUSION ICAM5 emerges as a promising biological marker with significant prognostic and therapeutic potential in LUAD.
Collapse
Affiliation(s)
- Yue Li
- Department of Respiratory Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, No. 26 of Heping Road, Xiangfang District, Harbin, 150040, China
| | - Keru Ma
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Hao Wang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zongying Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhuying Li
- Department of Respiratory Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, No. 26 of Heping Road, Xiangfang District, Harbin, 150040, China.
| |
Collapse
|
7
|
Cinnamon E, Stein I, Zino E, Rabinovich S, Shovman Y, Schlesinger Y, Salame TM, Reich-Zeliger S, Albrecht T, Roessler S, Schirmacher P, Lotem M, Ben-Neriah Y, Parnas O, Pikarsky E. RORc-expressing immune cells negatively regulate tertiary lymphoid structure formation and support their pro-tumorigenic functions. J Hepatol 2025; 82:1050-1067. [PMID: 39710149 DOI: 10.1016/j.jhep.2024.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 11/28/2024] [Accepted: 12/07/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND & AIMS RORc-expressing immune cells play important roles in inflammation, autoimmune disease and cancer. They are required for lymphoid organogenesis and have been implicated in tertiary lymphoid structure (TLS) formation. TLSs are formed in many cancer types and have been correlated with better prognosis and response to immunotherapy. In liver cancer, some TLSs are pro-tumorigenic as they harbor tumor progenitor cells and support their growth. The processes involved in TLS development and acquisition of pro- or anti-tumorigenic roles are largely unknown. This study aims to explore the role of RORc-expressing cells in TLS development in the context of inflammation-associated liver cancer. METHODS IKKβ(EE)Hep mice, exhibiting chronic liver inflammation, TLS formation and liver cancer, were crossed with RORc knockout mice to explore RORc's effect on TLS and tumor formation. TLS phenotypes were analyzed using transcriptional, proteomic, and immunohistochemical techniques. CD4, CD8, and B-cell depletions were used to assess their contribution to liver TLS and tumor formation. RESULTS RORc-expressing cells are detected within TLSs of both human patients and mice developing intrahepatic cholangiocarcinoma. In mice, these cells negatively regulate TLS formation, as excess TLSs form in their absence. CD4 cells are essential for liver TLS formation, while B cells are required for TLS formation specifically in the absence of RORc-expressing cells. Importantly, in chronically inflamed livers lacking RORc-expressing cells, TLSs become anti-tumorigenic, reducing tumor load. Anti-tumorigenic TLSs revealed enrichment of exhausted CD8 cells with effector functions, germinal center B cells and plasma cells. B cells are key in limiting tumor development, possibly via tumor-directed antibodies. CONCLUSIONS RORc-expressing cells negatively regulate B-cell responses and facilitate the pro-tumorigenic functions of hepatic TLSs. IMPACT AND IMPLICATIONS RORc-expressing immune cells play critical roles in immune regulation, yet their specific influence on tertiary lymphoid structures (TLSs) in liver pathology and cancer has not been elucidated. Our study reveals that RORc-expressing cells act as negative regulators of TLS formation and shape the immune microenvironment in a manner that promotes tumor development. In the absence of RORc-expressing cells, TLSs not only increase in number but also acquire anti-tumorigenic properties. These findings suggest that RORc-expressing cells serve as key modulators of liver immune dynamics, with potential implications for the use of RORc as a biomarker to differentiate between pro- and anti-tumorigenic immune environments and as a target for manipulating TLS abundance and phenotype in liver cancer.
Collapse
Affiliation(s)
- Einat Cinnamon
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Ilan Stein
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel; Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Elvira Zino
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Stav Rabinovich
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Yehuda Shovman
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel; Department of Neurology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Yehuda Schlesinger
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Tomer-Meir Salame
- Flow Cytometry Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | | | - Thomas Albrecht
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephanie Roessler
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michal Lotem
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Yinon Ben-Neriah
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Oren Parnas
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Eli Pikarsky
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel; Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
8
|
Zhao Z, Qiu S, Zhang X, Liu S, Wang L, Guan H, He J, Hu Y, Li X, Luo S, Chen Z, Mo T, Zhang Y, Zhao X, Pan Y, Ding H, Cao J, Pan J. Characterization of a novel cell line established from mice gastrointestinal stromal model by chemical induction. Transl Oncol 2025; 56:102388. [PMID: 40233502 PMCID: PMC12022689 DOI: 10.1016/j.tranon.2025.102388] [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: 09/23/2024] [Revised: 02/03/2025] [Accepted: 04/05/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are a type of tumor that originates from gastrointestinal mesenchymal tissue. Although several somatic or germline mutation GIST mice were established, however, there is still a lack of an authentic mice GIST cell lines for further experimental study. METHODS We developed a chemically induced C57BL/6 J GIST model using 3- methylcholanthrene. Tumor characteristics were confirmed through histology and IHC. Primary cells were isolated to establish the mGSTc01 cell line, and molecular profiling was conducted. Additionally, we established GIST model in immunocompetent mice to evaluate their sensitivity to imatinib. RESULTS Our study successfully developed a chemically induced murine GIST model, characterized by positive staining of c-kit and DOG-1. The mGSTc01 monoclonal cell line exhibited slender morphology and expressed the c-kit marker, Whole exome sequencing uncovered mutations of Lamb1, MMP9, and c-kit in GIST cells and provided a detailed picture of the entire genome's copy number variations. RNA sequencing indicated genes associated with cell adhesion and focal adhesion were enriched in mGSTc01 cells. The mGSTc01 cells demonstrated obvious malignant behaviors, notably elevated migration, adhesion, and proliferation. In immunocompetent mice, subcutaneous xenografts not only reserved the aggressive phenotype but also displayed a response to imatinib, underscoring the model's applicability for advancing therapeutic research. CONCLUSION We firstly established a mGSTc01 cell line derived from C57BL/6 J mice GIST tumor offers, which closely mimicking human disease characteristics. It is a potent platform for investigating tumor microenvironment of GIST in mice model, and provides a novel way for new therapeutic discoveries in GIST.
Collapse
Affiliation(s)
- Zhan Zhao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Shenghui Qiu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China; Department of General Surgery, Guangzhou First People's Hospital, Guangzhou, 510180, PR China
| | - Xiangwei Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Shijin Liu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Lu Wang
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, Guangzhou, Guangdong, 510632, PR China
| | - Hanyang Guan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Jiashuai He
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Yangzhi Hu
- The Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, PR China
| | - Xiaobo Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Simin Luo
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Zuyang Chen
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Tianmu Mo
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Yiran Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Xiaoxu Zhao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Yunlong Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Hui Ding
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China.
| | - Jie Cao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China; Department of General Surgery, Guangzhou First People's Hospital, Guangzhou, 510180, PR China.
| | - Jinghua Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China.
| |
Collapse
|
9
|
Yang J, Lu X, Cai Q, Liu M, Xia T, Hong D, Le L, Zhang X, Zhang X. Loss of TACC2 impairs chemokine CCL3 and CCL4 expression and reduces response to anti-PD-1 therapy in soft tissue sarcoma. Mol Cancer 2025; 24:158. [PMID: 40442694 PMCID: PMC12123857 DOI: 10.1186/s12943-025-02354-2] [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: 02/17/2025] [Accepted: 05/14/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Soft tissue sarcoma (STS) is a rare, heterogeneous malignancy with limited treatment options for metastatic disease. Despite advances in immunotherapy, including PD-1 inhibitors, clinical outcomes remain suboptimal, highlighting the need for novel biomarkers and therapeutic strategies. This study investigated the role of TACC2 in STS, focusing on its impact on the immune microenvironment and immunotherapy response. METHODS Whole-exome sequencing was performed to characterize TACC2-related genomic alterations in STS cohorts, complemented by immunohistochemistry for protein-level validation. Mechanistic insights were obtained through chromatin immunoprecipitation (ChIP) and co-immunoprecipitation assays, focusing on TACC2's interaction with the NuRD/CoREST complex. The efficacy of anti-PD-1 therapy was evaluated in TACC2-overexpressing mouse models, and clinical relevance was analyzed using patient survival and treatment response data. RESULTS TACC2 acted as a tumor suppressor in STS, with low expression associated with poor overall survival. Mechanistically, TACC2 enhanced CCL3 and CCL4 transcription, promoting CD8 + T cell infiltration by inhibiting NuRD/CoREST nuclear translocation. In vivo, TACC2 overexpression synergized with PD-1 blockade therapy, leading to a significant reduction in tumor volume and prolonged survival. Clinically, high TACC2 expression was associated with improved responses to immunotherapy. CONCLUSIONS In conclusion, TACC2 is an important regulator of the immune response in STS, functioning as a tumor suppressor and a modulator of response to PD-1 blockade. Its role in modulating chemokine expression and CD8 + T cell infiltration highlights its potential as a therapeutic target and predictive biomarker for STS immunotherapy.
Collapse
Affiliation(s)
- Jing Yang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Xiuxia Lu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P.R. China
| | - Qiyan Cai
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Mengmeng Liu
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Jiangxi Key Laboratory of Clinical Translational Cancer Research, Nanchang University, Nanchang, Jiangxi, 330000, P.R. China
| | - Tianliang Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Dongchun Hong
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Liyuan Le
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Xinke Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xing Zhang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
| |
Collapse
|
10
|
Hegoburu A, Amer M, Frizelle F, Purcell R. B cells and tertiary lymphoid structures in cancer therapy response. BJC REPORTS 2025; 3:40. [PMID: 40437260 PMCID: PMC12119954 DOI: 10.1038/s44276-025-00146-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 03/31/2025] [Accepted: 04/18/2025] [Indexed: 06/01/2025]
Abstract
Recent advances in immuno-oncology research have revolutionised our understanding of the interplay between immune cells and the tumour microenvironment (TME), profoundly impacting patient responses to therapy. The TME, comprising tumour cells, immune cells, extracellular matrix, stromal cells, and co-existing microbes, orchestrates the immune phenotype of cancers, shaping disease progression and treatment outcomes. Immune-cell infiltration serves as a significant prognostic marker in various cancers, with higher rates correlating with improved prognosis. Recent discoveries have paved the way for immune checkpoint blockade therapies, which exhibit remarkable efficacy across multiple cancer types. However, understanding the nuanced contributions of different immune-cell populations to therapeutic responses remains a challenge. The majority of research has focussed on the role of T cells in the immune response to cancer therapies, with the potential importance of B cells only recently being recognised. Here, we review the diverse phenotypes of B cells within the TME, their structural organisation within tertiary lymphoid structures (TLS), and the role of both B cells and TLS in cancer prognosis and response to different therapies for cancer treatment.
Collapse
Affiliation(s)
- Adèle Hegoburu
- Department of Surgery and Critical Care, Ōtākou Whakaihu Waka/University of Otago, Christchurch, Aotearoa New Zealand
| | - Mohammad Amer
- Department of Surgery and Critical Care, Ōtākou Whakaihu Waka/University of Otago, Christchurch, Aotearoa New Zealand
| | - Frank Frizelle
- Department of Surgery and Critical Care, Ōtākou Whakaihu Waka/University of Otago, Christchurch, Aotearoa New Zealand
| | - Rachel Purcell
- Department of Surgery and Critical Care, Ōtākou Whakaihu Waka/University of Otago, Christchurch, Aotearoa New Zealand.
| |
Collapse
|
11
|
Gallois C, Sroussi M, André T, Mouillet-Richard S, Agueeff N, Mulot C, Vernerey D, Louvet C, Bachet JB, Dourthe LM, Mazard T, Jary M, Coutzac C, Lecaille C, Tabernero J, Van Laethem JL, Lepage C, Emile JF, de Reyniès A, Taieb J, Laurent-Puig P. Prognostic Models From Transcriptomic Signatures of the Tumor Microenvironment and Cell Cycle in Stage III Colon Cancer From PETACC-8 and IDEA-France Trials. J Clin Oncol 2025; 43:1765-1776. [PMID: 39889251 PMCID: PMC12084023 DOI: 10.1200/jco.23.02262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/08/2024] [Accepted: 12/23/2024] [Indexed: 02/02/2025] Open
Abstract
PURPOSE The objective of this work was to establish prognostic models in stage III colon cancer (CC) on the basis of transcriptomic signatures of the tumor microenvironment (TME) and cell cycle from the PETACC-8 (training set) and IDEA-France (validation set) trials. PATIENTS AND METHODS 3'RNA sequencing was performed in 1,733 patients from the PETACC-8 trial and 1,248 patients from the IDEA-France trial. Four transcriptomic signatures were analyzed: T-cell and macrophage M2 signatures, the expression of CXCL13, and a score on the basis of the Oncotype DX CC Recurrence Score using the same formula from the stromal score and the cell cycle score. The Immune Proliferative Stromal (IPS) score was defined as the number of dichotomized signatures that fall under the category of a dismal prognosis (from 0 to 4). Time to recurrence (TTR) was defined as the time from the date of random assignment to local and/or metastatic relapse and/or death because of CC, whichever occurs first. RESULTS High Oncotype-like and M2 scores and low CXCL13 expression and T-cell score were associated with a shorter TTR. A multivariable model including these signatures and all known prognostic factors applied to the IDEA-France cohort by obtaining a value of this model for each patient showed TTR significantly different depending on the quartile of this value and a 3-year rate of patients without recurrence ranging from 56% for the lowest quartile to 89% for the highest quartile (P < .0001). The IPS score was significantly associated with TTR in multivariable analysis. CONCLUSION Using transcriptomic data of patients with stage III CC from two large-scale adjuvant trials, a prognostic model on the basis of signatures of the TME and the cell cycle provides important information in addition to known prognostic factors for patient stratification on risk of recurrence.
Collapse
Affiliation(s)
- Claire Gallois
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
- Digestive Oncology Department, Institut du Cancer Paris CARPEM, APHP, APHP Centre, Hôpital Européen G. Pompidou, Université Paris Cité, Paris, France
| | - Marine Sroussi
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
- Institut Chimie Biologie Innovation—Laboratoire de BioChimie, ESPCI, UMR8231 CNRS, Université PSL, Paris, France
| | - Thierry André
- Department of Medical Oncology, Hôpital Saint Antoine, Sorbonne Université, Paris, France
| | - Sophie Mouillet-Richard
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
| | - Natacha Agueeff
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
| | - Claire Mulot
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
| | - Dewi Vernerey
- Methodology and Quality of Life Unit in Oncology, University of Besançon, Besançon, France
- Etablissement Français du Sang Bourgogne Franche-Comté, INSERM, Unité Mixte de Recherche 1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Franche-Comté University, Besançon University Hospital, Besançon, France
| | - Christophe Louvet
- Department of Medical Oncology, Institut Mutualiste Montsouris, Paris, France
| | - Jean-Baptiste Bachet
- Department of Hepato-Gastroenterology and Digestive Oncology, Pitié-Salpêtriére Hospital, APHP, Sorbonne Université, Paris, France
| | | | - Thibault Mazard
- Department of Medical Oncology, IRCM, INSERM, ICM, University of Montpellier, Montpellier, France
| | - Marine Jary
- Department of Digestive and Hepatobiliary Surgery, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Clélia Coutzac
- Department of Medical Oncology, Centre Leon Berard, University Claude Bernard Lyon, Lyon, France
| | - Cédric Lecaille
- Cancerology Department, Bordeaux Nord Polyclinic, Bordeaux, France
| | - Josep Tabernero
- Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | - Jean-Luc Van Laethem
- Department of Digestive Oncology, Hopital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Côme Lepage
- Hepatogastroenterology and Digestive Oncology Department, Dijon Bourgogne Hospital, University of Burgundy and Franche Comté, Dijon, France
| | - Jean-François Emile
- EA4340 BECCOH, Service de Pathologie, Hôpital Ambroise Paré, AP-HP, Université de Versailles SQY, Boulogne, France
| | - Aurélien de Reyniès
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
- Laboratoire SeqOIA, Paris, France
| | - Julien Taieb
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
- Digestive Oncology Department, Institut du Cancer Paris CARPEM, APHP, APHP Centre, Hôpital Européen G. Pompidou, Université Paris Cité, Paris, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Paris, France
| |
Collapse
|
12
|
Zaidi N, Jaffee EM, Yarchoan M. Recent advances in therapeutic cancer vaccines. Nat Rev Cancer 2025:10.1038/s41568-025-00820-z. [PMID: 40379970 DOI: 10.1038/s41568-025-00820-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/19/2025]
Abstract
The success of cancer prevention vaccines targeting cancer-causing viruses has drastically reduced cancer mortality worldwide. However, the development of therapeutic cancer vaccines, which aim to elicit an immune response directly against cancer cells, has faced notable clinical setbacks. In this Review, we explore lessons learned from past cancer vaccine trials and how the field has progressed into an era of renewed promise. Previous vaccines primarily targeted tumour-associated antigens and were mainly tested as monotherapies in late-stage cancers. In contrast, contemporary vaccines focus on targeting tumour-specific antigens (neoantigens) and are showing initial evidence of clinical efficacy, particularly in early-stage cancers and precancers when combined with immune checkpoint inhibitors. Advances in tumour profiling and novel vaccine platforms have enhanced vaccine specificity and potency. We discuss recent clinical trials of therapeutic cancer vaccines and outline future directions for the field.
Collapse
Affiliation(s)
- Neeha Zaidi
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Elizabeth M Jaffee
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| | - Mark Yarchoan
- Johns Hopkins Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
13
|
Rahal Z, El Darzi R, Moghaddam SJ, Cascone T, Kadara H. Tumour and microenvironment crosstalk in NSCLC progression and response to therapy. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01021-1. [PMID: 40379986 DOI: 10.1038/s41571-025-01021-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 05/19/2025]
Abstract
The treatment landscape of non-small-cell lung cancer (NSCLC) is evolving rapidly, driven by advances in the development of targeted agents and immunotherapies. Despite this progress, some patients have suboptimal responses to treatment, highlighting the need for new therapeutic strategies. In the past decade, the important role of the tumour microenvironment (TME) in NSCLC progression, metastatic dissemination and response to treatment has become increasingly evident. Understanding the complexity of the TME and its interactions with NSCLC can propel efforts to improve current treatment modalities, overcome resistance and develop new treatments, which will ultimately improve the outcomes of patients. In this Review, we provide a comprehensive view of the NSCLC TME, examining its components and highlighting distinct archetypes characterized by spatial niches within and surrounding tumour nests, which form complex neighbourhoods. Next, we explore the interactions within these components, focusing on how inflammation and immunosuppression shape the dynamics of the NSCLC TME. We also address the emerging influences of patient-related factors, such as ageing, sex and health disparities, on the NSCLC-TME crosstalk. Finally, we discuss how various therapeutic strategies interact with and are influenced by the TME in NSCLC. Overall, we emphasize the interconnectedness of these elements and how they influence therapeutic outcomes and tumour progression.
Collapse
Affiliation(s)
- Zahraa Rahal
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Roy El Darzi
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina Cascone
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
14
|
Li Y, Bhargava R, Tran JT, Blane TR, Peng L, Luan F, Huang Z, Zhang Z, Sun Y, Xiao C, Nemazee D. Blocking plasma cell fate enhances antigen-specific presentation by B cells to boost anti-tumor immunity. Nat Commun 2025; 16:4454. [PMID: 40360528 PMCID: PMC12075458 DOI: 10.1038/s41467-025-59622-4] [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: 08/28/2024] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
B cells engage in anti-tumor immunity but how they contribute to cancer suppression remains unclear. We report that inhibiting plasma cell differentiation either in IgMi mice lacking Igh elements needed for antibody secretion or in mice with B cell-specific knockout of Blimp-1 (Blimp-1 BcKO) promotes rather than inhibits antitumor immunity and increases numbers of activated B cells. Deficiency of Blimp-1 in tumor-infiltrating B cells generates a unique transcription profile associated with expansion of mutated clones targeting cognate tumor cells. Major histocompatibility complex class II (MHC II) is required for anti-tumor efficacy. Blimp-1-deficient B cells have increased expression of CD80 and CD86 costimulatory molecules that enhance effector T cell function. The Blimp-1 inhibitor valproic acid suppresses tumor growth in a B cell-dependent manner. Thus, inhibition of plasma cell differentiation results in enhanced tumor-specific antigen presentation by B cells and thereby tumor repression, suggesting a potential avenue of immunotherapy against cancer.
Collapse
Affiliation(s)
- Yunqiao Li
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Raag Bhargava
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Jenny Tuyet Tran
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Tanya R Blane
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Linghang Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Fangkun Luan
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Zhe Huang
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Zefan Zhang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Yunfan Sun
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Changchun Xiao
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| | - David Nemazee
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
15
|
Shu L, Tao T, Xiao D, Liu S, Tao Y. The role of B cell immunity in lung adenocarcinoma. Genes Immun 2025:10.1038/s41435-025-00331-9. [PMID: 40360749 DOI: 10.1038/s41435-025-00331-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 04/07/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
Lung cancer is the deadliest cancer globally. Non-small cell lung cancer (NSCLC), including adenocarcinoma, squamous cell carcinoma, and large cell carcinoma, constitutes a significant portion of cases. Adenocarcinoma, the most prevalent type, has seen a rising incidence. Immune checkpoint inhibitors (ICIs) have improved outcomes in lung adenocarcinoma (LUAD), yet response rates remain unsatisfactory. PD-1/PD-L1 inhibitors are primary ICIs for LUAD, targeting the PD-1/PD-L1 pathway between CD8+ T cells and tumor cells. However, LUAD presents a "cold tumor" phenotype with fewer CD8+ T cells and lower PD-1 expression, leading to resistance to ICIs. Thus, understanding the function of other immune cell in tumor microenvironment is crucial for developing novel immunotherapies for LUAD. B cells, which is part of the adaptive immune system, have gained attention for its role in cancer immunology. While research on B cells lags behind T cells, recent studies reveal their close correlation with prognosis and immunotherapy effectiveness in various solid tumors, including lung cancer. B cells show higher abundance, activity, and prognostic significance in LUAD than that in LUSC. This review summarizes the difference of B cell immunity between LUAD and other lung cancers, outlines the role of B cell immunity in LUAD.
Collapse
Affiliation(s)
- Long Shu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Tania Tao
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, School of Basic Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
16
|
Li H, Zhang MJ, Zhang B, Lin WP, Li SJ, Xiong D, Wang Q, Wang WD, Yang QC, Huang CF, Deng WW, Sun ZJ. Mature tertiary lymphoid structures evoke intra-tumoral T and B cell responses via progenitor exhausted CD4 + T cells in head and neck cancer. Nat Commun 2025; 16:4228. [PMID: 40335494 PMCID: PMC12059173 DOI: 10.1038/s41467-025-59341-w] [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: 06/08/2024] [Accepted: 04/18/2025] [Indexed: 05/09/2025] Open
Abstract
Tumor tertiary lymphoid structures (TLS), especially mature TLS (mTLS), have been associated with better prognosis and improved responses to immune checkpoint blockade (ICB), but the underlying mechanisms remain incompletely understood. Here, by performing single-cell RNA, antigen receptor sequencing and spatial transcriptomics on tumor tissue from head and neck squamous cell carcinoma (HNSCC) patients with different statuses of TLS, we observe that mTLS are enriched with stem-like T cells, and B cells at various maturation stages. Notably, progenitor exhausted CD4+ T cells, with features resembling follicular helper T cells, support these responses, by activating B cells to produce plasma cells in the germinal center, and interacting with DC-LAMP+ dendritic cells to support CD8+ T cell activation. Conversely, non-mTLS tumors do not promote local anti-tumor immunity which is abundant of immunosuppressive cells or a lack of stem-like B and T cells. Furthermore, patients with mTLS manifest improved overall survival and response to ICB compared to those with non-mTLS. Overall, our study provides insights into mechanisms underlying mTLS-mediated intra-tumoral immunity events against cancer.
Collapse
Affiliation(s)
- Hao Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Department of Oral Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Meng-Jie Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Boxin Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Wen-Ping Lin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Shu-Jin Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Dian Xiong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Qing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Wen-Da Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Qi-Chao Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Cong-Fa Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Wei-Wei Deng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
- Department of Oral Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
- Department of Oral Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
17
|
Chen Y, Wu Y, Zhao Z, Wen L, Wu M, Song D, Zeng Q, Liu Y, Yan G, Zhang G. Retrospective study on the correlation between CXCL13, immune infiltration, and tertiary lymphoid structures in cutaneous squamous cell carcinoma. PeerJ 2025; 13:e19398. [PMID: 40352278 PMCID: PMC12065455 DOI: 10.7717/peerj.19398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Background C-X-C motif chemokine ligand 13 (CXCL13) is a crucial chemokine for the recruitment of immune cells and the formation of tertiary lymphoid structure (TLS) in the tumor microenvironment. However, the relationship between CXCL13 and immune infiltration in cutaneous squamous cell carcinoma (cSCC) remains unclear. Objective We aimed to investigate the expression of CXCL13 and explore its association with immune activation and TLS in cSCC. Methods A total of 63 cSCC patients were involved in the present study. Hematoxylin and eosin staining was used for pathological examination of cSCC. Bioinformatics analyses and immunohistochemical staining were employed to access the expression of CXCL13 and TLS states. Public single cell RNA-sequencing atlas of skin disorders and multiplex immunofluorescence were used to explore CXCL13-producing cells. Results Utilizing the public database and our clinical cohort, we observed robust CXCL13 expression in cSCC tissues and a significant correlation with immune activation. Higher expression levels of CXCL13 were associated with lower histopathological grades and increased TLS formation. Furthermore, we confirmed that T cells and fibroblasts were the predominant cell types of CXCL13 secretion in cSCC. Conclusions CXCL13 is up-regulated in cSCC, which shows a significant positive correlation with immune infiltration and TLS formation. Our results underscore the role of CXCL13 in shaping the cSCC microenvironment, highlighting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yulu Chen
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuhao Wu
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zijun Zhao
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Long Wen
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Mingshun Wu
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dekun Song
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingyu Zeng
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yeqiang Liu
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guorong Yan
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guolong Zhang
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
18
|
Soupir A, Ospina OE, Hampton O, Churchman M, Radmacher M, Hedges D, McKean D, Agius P, Zeeshan S, Seligson ND, Pollock R, Liebner D, Chen JL, Tinoco G, Salhia B, McCarter M, Wilky BA, Miller BJ, Cavnar MJ, Groundland JS, Schneider BP, Riedlinger G, Edge SB, Moskaluk CA, Cardona K, Naqash AR, Gonzalez RJ, Mullinax JE, Joyce DM, Binitie O, Douglas Letson G, Naghavi AO, Druta M, Reed DR, Siegel EM, Teer JK, Fridley BL, Brohl AS. Genomic, transcriptomic, and immunogenomic landscape of over 1300 sarcomas of diverse histology subtypes. Nat Commun 2025; 16:4206. [PMID: 40328759 PMCID: PMC12055966 DOI: 10.1038/s41467-025-58678-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Given their rarity and diversity, a fundamental understanding of the genomic underpinnings for many sarcoma subtypes is still lacking. To better define the molecular landscape of this group of diseases, we perform matched whole exome sequencing and RNA sequencing on a cohort of 1340 sarcoma tumor specimens. We identify recurrent somatic mutations and observe an increased mutational burden in metastatic vs. primary samples (p < 0.001). We observe frequent copy number alterations including whole genome doubling, with this feature being more common in metastatic tumors (p = 0.026). Estimation of immune cell abundances followed by hierarchical clustering identifies five immune subtypes ranging from low to high and we observe inferior overall survival in immune deplete clusters compared to immune enriched (p < 0.01). Interestingly, GIST predominantly form a distinct "immune intermediate" cluster that is marked by a specific enrichment for NK cells (FDR < 0.01).
Collapse
Affiliation(s)
- Alex Soupir
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Oscar E Ospina
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | | | | | | | | | | | | | - Saman Zeeshan
- Department of Biomedical and Health Informatics, School of Medicine, University of Missouri, Kansas City, MO, USA
| | - Nathan D Seligson
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
| | - Raphael Pollock
- Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - David Liebner
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - James L Chen
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Gabriel Tinoco
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bodour Salhia
- Department of Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | | | - Benjamin J Miller
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IO, USA
| | - Michael J Cavnar
- Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - John S Groundland
- University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Bryan P Schneider
- Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA
| | | | - Stephen B Edge
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Kenneth Cardona
- Division of Surgical Oncology, Emory University, Atlanta, GA, USA
| | - Abdul Rafeh Naqash
- Medical Oncology/Phase 1 program, Stephenson Cancer Center, University of Oklahoma Health Sciences, Oklahoma City, OK, USA
| | | | | | - David M Joyce
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, USA
| | - Odion Binitie
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Arash O Naghavi
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Mihaela Druta
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, USA
| | - Damon R Reed
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, FL, USA
| | - Erin M Siegel
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Brooke L Fridley
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
- Division of Health Services and Outcomes Research, Children's Mercy, Kansas City, MO, USA
| | - Andrew S Brohl
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
19
|
Che Y, Lee J, Abou-Taleb F, Rieger KE, Satpathy AT, Chang ALS, Chang HY. Induced B cell receptor diversity predicts PD-1 blockade immunotherapy response. Proc Natl Acad Sci U S A 2025; 122:e2501269122. [PMID: 40314973 PMCID: PMC12067265 DOI: 10.1073/pnas.2501269122] [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: 01/17/2025] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Immune checkpoint inhibitors such as anti-Programmed Death-1 antibodies (aPD-1) can be effective in treating advanced cancers. However, many patients do not respond, and the mechanisms underlying these differences remain incompletely understood. In this study, we profile a cohort of patients with locally advanced or metastatic basal cell carcinoma undergoing aPD-1 therapy using single-cell RNA sequencing, high-definition spatial transcriptomics in tumors and draining lymph nodes, and spatial immunoreceptor profiling, with long-term clinical follow-up. We find that successful responses to PD-1 inhibition are characterized by an induction of B cell receptor (BCR) clonal diversity after treatment initiation. These induced BCR clones spatially colocalize with T cell clones, facilitate their activation, and traffic alongside them between tumor and draining lymph nodes to enhance tumor clearance. Furthermore, we validated aPD-1-induced BCR diversity as a predictor of clinical response in a larger cohort of glioblastoma, melanoma, and head and neck squamous cell carcinoma patients, suggesting that this is a generalizable predictor of treatment response across many types of cancers. We find that pretreatment tumors harbor a characteristic gene expression signature that portends a higher probability of inducing BCR clonal diversity after aPD-1 therapy, and we develop a machine learning model that predicts PD-1-induced BCR clonal diversity from baseline tumor RNA sequencing. These findings underscore a dynamic role of B cell diversity during immunotherapy, highlighting its importance as a prognostic marker and a potential target for intervention in non-responders.
Collapse
Affiliation(s)
- Yonglu Che
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA94063
| | - Jinwoo Lee
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA94063
| | - Farah Abou-Taleb
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA94063
| | - Kerri E. Rieger
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA94063
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94304
| | - Ansuman T. Satpathy
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94304
| | - Anne Lynn S. Chang
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA94063
| | - Howard Y. Chang
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA94063
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94304
- Department of Genetics, Stanford University School of Medicine, Stanford, CA94305
- HHMI, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
20
|
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
|
21
|
Gorvel L, Panouillot M, Rouvière MS, Billon E, Fattori S, Sonongbua J, Boucherit N, Ben Amara A, Quilichini O, Granjeaud S, Degos C, Nunès JA, Carcopino X, Lambaudie E, Chrétien AS, Sabatier R, Dieu-Nosjean MC, Olive D. Tertiary Lymphoid Structures Are Associated with Enhanced Macrophage Activation and Immune Checkpoint Expression and Predict Outcome in Cervical Cancer. Cancer Immunol Res 2025; 13:712-728. [PMID: 39888676 DOI: 10.1158/2326-6066.cir-24-0979] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/23/2024] [Accepted: 01/31/2025] [Indexed: 02/02/2025]
Abstract
Cervical tumors are usually treated using surgery, chemotherapy, and radiotherapy and would benefit from immunotherapies. However, the immune microenvironment in cervical cancer remains poorly described. Tertiary lymphoid structures (TLS) were recently described as markers for better immunotherapy response and overall better prognosis in patients with cancer. We evaluated the cervical tumor immune microenvironment, specifically focusing on TLS, using combined high-throughput phenotyping, soluble factor concentration dosage in the tumor microenvironment, and spatial interaction analyses. We found that TLS presence was associated with a more inflammatory soluble microenvironment, with the presence of B cells as well as more activated macrophages and dendritic cells (DC). Furthermore, this myeloid cell activation was associated with the expression of immune checkpoints, such as PD-L1 and CD40, and the proximity of activated conventional type 2 DCs to CD8+ T cells, indicating better immune interactions and tumor control. Finally, we associated TLS presence, greater B-cell density, and activated DC density with improved progression-free survival, substantiating TLS presence as a potential prognostic marker. Our results provide evidence that TLS presence denotes cell activation and immunotherapy target expression.
Collapse
Affiliation(s)
- Laurent Gorvel
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Marylou Panouillot
- UMRS1135 Sorbonne University, INSERM U1135, Centre of Immunology and Microbial Infections (Cimi), Immune Microenvironment and Immunotherapy Team, Paris, France
| | - Marie-Sarah Rouvière
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Emilien Billon
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Stéphane Fattori
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Jumaporn Sonongbua
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Nicolas Boucherit
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Amira Ben Amara
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Olivia Quilichini
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Samuel Granjeaud
- CRCM Integrative Bioinformatics Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Clara Degos
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Jacques A Nunès
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Xavier Carcopino
- Department of Obstetrics and Gynecology, Hôpital Nord, APHM, Aix-Marseille University (AMU), CNRS, IRD, IMBE UMR 7263, 13397, Marseille, France
| | - Eric Lambaudie
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Anne-Sophie Chrétien
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Renaud Sabatier
- Predictive Oncology Laboratory, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Marie-Caroline Dieu-Nosjean
- UMRS1135 Sorbonne University, INSERM U1135, Centre of Immunology and Microbial Infections (Cimi), Immune Microenvironment and Immunotherapy Team, Paris, France
| | - Daniel Olive
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| |
Collapse
|
22
|
Yamamoto Y, Nonomura N. Editorial Comment to "Tertiary Lymphoid Structures Correlate With Better Prognosis in Patients With Retroperitoneal Sarcoma: A Retrospective Study". Int J Urol 2025. [PMID: 40312933 DOI: 10.1111/iju.70092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Affiliation(s)
- Yoshiyuki Yamamoto
- Department of Urology, The University of Osaka Graduate School of Medicine, Suita City, Osaka Prefecture, Japan
| | - Norio Nonomura
- Department of Urology, The University of Osaka Graduate School of Medicine, Suita City, Osaka Prefecture, Japan
| |
Collapse
|
23
|
Kret ZS, Sweder RJ, Pollock R, Tinoco G. Potential Mechanisms for Immunotherapy Resistance in Adult Soft-Tissue Sarcoma. Target Oncol 2025; 20:485-502. [PMID: 40289241 DOI: 10.1007/s11523-025-01145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2025] [Indexed: 04/30/2025]
Abstract
Soft-tissue sarcomas represent a diverse group of rare malignancies originating from mesenchymal tissue, accounting for less than 1% of adult cancers in the USA. With over 13,000 new cases and around 5350 deaths annually, patients with metastatic soft-tissue sarcomas face limited therapeutic options and an estimated median overall survival of 18 months. While immunotherapy has demonstrated effectiveness in several cancers, its application in soft-tissue sarcomas remains challenging owing to the tumors' largely "cold" immunological environment, characterized by low levels of tumor-infiltrating lymphocytes and a lack of soft-tissue sarcoma-specific biomarkers. This review examines potential mechanisms underlying immunotherapy resistance in soft-tissue sarcomas, including the complex interplay between innate and adaptive immunity, the tumor microenvironment, and the role of immune-related genes. Despite preliminary findings suggesting correlations between immune profiles and histological subtypes, consistent biomarkers for predicting immunotherapeutic responses across soft-tissue sarcoma types are absent. Emerging strategies focus on converting "cold" tumors to "hot" tumors, enhancing their susceptibility to immunologic activation. While research is ongoing, personalized treatment approaches may offer hope for overcoming the inherent heterogeneity and resistance seen in soft-tissue sarcomas, ultimately aiming to improve outcomes for affected patients.
Collapse
Affiliation(s)
- Zaina S Kret
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ryan J Sweder
- The Ohio State University College of Arts and Sciences and College of Medicine, Columbus, OH, USA
| | - Raphael Pollock
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Gabriel Tinoco
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 1800 Cannon Drive, 1240 Lincoln Tower, Columbus, OH, 43210, USA.
| |
Collapse
|
24
|
Zhang P, Tao C, Xie H, Yang L, Lu Y, Xi Y, Yao S, Yuan L, Guo P, Cheng X. Identification of CD66c as a potential target in gastroesophageal junction cancer for antibody-drug conjugate development. Gastric Cancer 2025; 28:422-441. [PMID: 39918687 PMCID: PMC11993476 DOI: 10.1007/s10120-025-01584-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 12/27/2024] [Indexed: 04/13/2025]
Abstract
BACKGROUND Gastroesophageal junction (GEJ) cancer exhibits unique biological characteristics and currently lacks specific targeted therapies. Given the clinical efficacy of antibody-drug conjugates (ADCs) in solid tumor treatment, we aimed to identify a novel ADC target and suitable payload for GEJ-targeted therapy. METHODS In this study, we conducted bioinformatic analyses of multi-omics data, including transcriptomics, proteomics, and phosphoproteomics, to identify CD66c as a promising ADC target for GEJ cancer. We then engineered a CD66c-directed antibody-drug conjugate (CD66c-DXd) incorporating a GGFG linker. The preclinical efficacy of CD66c-DXd was determined in multi GEJ xenograft models. RESULTS Proteomic analyses of 103 cases of GEJ cancer revealed that CD66c expression was significantly higher in tumoral tissues compared to normal tissues. Proteomic and phosphoproteomic analyses identified deruxtecan (DXd) as a potentially potent payload for ADCs targeting GEJ cancer. Furthermore, high CD66c expression in GEJ was associated with a significantly lower proportion of plasma cells. The drug-to-antibody ratio (DAR) of CD66c-DXd was determined to be 3.6. CD66c-DXd effectively and selectively ablated multiple human GEJ cell lines (OE-19, OE33 and SK-GT-4) without affecting non-malignant cells (GES-1) in vitro. Eventually, CD66c-DXd mediated potent and durable tumor regression in vivo with excellent safety profiles. CONCLUSIONS This preclinical study provides a strong rationale for the further development of CD66c-DXd as promising therapeutic candidates to treat advanced GEJ cancer. Additionally, the study demonstrates the robustness of the multi-omics data in identifying novel potential ADC targets and payloads.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Department of Medical Oncology, Zhejiang Provincial People's Hospital, Hangzhou, 310022, Zhejiang, China
| | - Changjuan Tao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Hanfei Xie
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Postgraduate Training Base Alliance of Wenzhou Medical University, Hangzhou, 310022, Zhejiang, China
| | - Liu Yang
- Department of Medical Oncology, Zhejiang Provincial People's Hospital, Hangzhou, 310022, Zhejiang, China
| | - Ye Lu
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yun Xi
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Shili Yao
- School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, China
| | - Li Yuan
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Peng Guo
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China.
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China.
| |
Collapse
|
25
|
Bindu S, Bibi R, Pradeep R, Sarkar K. The evolving role of B cells in malignancies. Hum Immunol 2025; 86:111301. [PMID: 40132250 DOI: 10.1016/j.humimm.2025.111301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
B cells play diverse roles in different pathological circumstances, such as neoplastic diseases, autoimmune disorders, and neurological maladies. B cells, which are essential elements of the adaptive immune system, demonstrate exceptional functional variety, including the generation of antibodies, the presentation of antigens, and the secretion of cytokines. Within the field of oncology, B cells display a multifaceted nature in the tumor microenvironment, simultaneously manifesting both tumor-promoting and tumor-suppressing characteristics. Studies have found that the existence of tertiary lymphoid structures, which consist of B cells, is linked to better survival rates in different types of cancers. This article examines the involvement of B cells in different types of malignancies, emphasizing their importance in the development of the diseases and their potential as biomarkers. Additionally, the review also examines the crucial role of B cells in autoimmune illnesses and their potential as targets for therapy. The article also analyses the role of B cells in immunization and exploring their potential uses in cancer immunotherapy. This analysis highlights the intricate and occasionally contradictory roles of B cells, underlining the necessity for additional research to clarify their varied actions in various illness scenarios.
Collapse
Affiliation(s)
- Soham Bindu
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - Roshni Bibi
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - R Pradeep
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India.
| |
Collapse
|
26
|
Nagase Y, Kodama M, Aimono E, Nakamura K, Takamatsu R, Abe K, Yoshimura T, Chiyoda T, Yamagami W, Nishihara H. CXCL9 and CXCL13 shape endometrial cancer immune-activated microenvironment via tertiary lymphoid structure formation. Cancer Sci 2025; 116:1193-1202. [PMID: 39960836 PMCID: PMC12044659 DOI: 10.1111/cas.16371] [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: 06/18/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 05/02/2025] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has been successfully applied to various cancers; however, not all patients respond to ICI therapy. Tumors with an immune-activated environment are highly responsive to ICIs. To identify the cells and molecules essential to the formation of an immune-activated cancer microenvironment, we focused on the tertiary lymphoid structure (TLS) and performed histological and genomic analyses using endometrial cancer material. In the high immunogenic group, numerous TLSs were observed, and CXCL9 and CXCL13 expression was markedly increased. CXCL9-positive antigen-presenting and CXCL13-positive follicular dendritic cells were distributed in the T- and B-cell zones of TLSs, respectively. A group of molecules whose expression was upregulated along with CXCL9 and CXCL13 expression was strongly associated with cellular immunity. These results suggest that CXCL9-expressing antigen-presenting cells and CXCL13-expressing follicular dendritic cells coordinately shape the immune-activated microenvironment through TLS formation. The current findings will contribute to a better understanding of the mechanisms underlying the activated cancer immune microenvironment, thereby advancing the field of precision cancer medicine.
Collapse
Affiliation(s)
| | - Makoto Kodama
- Department of PathologyTokyo Yamate Medical CenterTokyoJapan
- Department of Human Pathology, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Eriko Aimono
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Reika Takamatsu
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Keiko Abe
- Department of PathologyTokyo Yamate Medical CenterTokyoJapan
| | - Takuma Yoshimura
- Department of Obstetrics and GynecologyKeio University School of MedicineTokyoJapan
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and GynecologyKeio University School of MedicineTokyoJapan
| | - Wataru Yamagami
- Department of Obstetrics and GynecologyKeio University School of MedicineTokyoJapan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| |
Collapse
|
27
|
He B, Wood KH, Li ZJ, Ermer JA, Li J, Bastow ER, Sakaram S, Darcy PK, Spalding LJ, Redfern CT, Canes J, Oliveira M, Prat A, Cortes J, Thompson EW, Littlefield BA, Redfern A, Ganss R. Selective tubulin-binding drugs induce pericyte phenotype switching and anti-cancer immunity. EMBO Mol Med 2025; 17:1071-1100. [PMID: 40140727 DOI: 10.1038/s44321-025-00222-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
The intratumoral immune milieu is crucial for the success of anti-cancer immunotherapy. We show here that stromal modulation by the tubulin-binding anti-cancer drugs combretastatin A4 (CA-4) and eribulin improved tumor perfusion and anti-tumor immunity. This was achieved by reverting highly proliferative, angiogenic pericytes into a quiescent, contractile state which durably normalized the vascular bed and reduced hypoxia in mouse models of pancreatic neuroendocrine cancer, breast cancer and melanoma. The crucial event in pericyte phenotype switching was RhoA kinase activation, which distinguished CA-4 and eribulin effects from other anti-mitotic drugs such as paclitaxel and vinorelbine. Importantly, eribulin pre-treatment sensitized tumors for adoptive T cell therapy or checkpoint inhibition resulting in effector cell infiltration and better survival outcomes in mice. In breast cancer patients, eribulin neoadjuvant treatment induced pericyte maturity and RhoA kinase activity indicating similar vessel remodeling effects as seen in mice. Moreover, a contractile pericyte signature was associated with overall better survival outcome in two independent breast cancer cohorts. This underscores the potential of re-purposing specific anti-cancer drugs to enable synergistic complementation with emerging immunotherapies.
Collapse
Affiliation(s)
- Bo He
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Kira H Wood
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Zhi-Jie Li
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Guangdong, P. R. China
| | - Judith A Ermer
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Ji Li
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Edward R Bastow
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | | | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Lisa J Spalding
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Cameron T Redfern
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Jordi Canes
- SOLTI Cancer Research Group, Barcelona, Spain
| | - Mafalda Oliveira
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Aleix Prat
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Javier Cortes
- SOLTI Cancer Research Group, Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR)-Oncoclínicas&Co, Jersey City, NJ, USA
- Medica Scientia Innovation Research (MEDSIR)-Oncoclínicas&Co, Sao Paulo, Brazil
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
- IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid, Spain
| | - Erik W Thompson
- School of Biomedical Sciences and Centre for Genomics and Personalised Health, Faculty of Health, Queensland University of Technology (QUT) and Translational Research Institute, Brisbane, Australia
| | | | - Andrew Redfern
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- Fiona Stanley Hospital, Perth, WA, Australia
| | - Ruth Ganss
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
28
|
Avallone G, Brigandì E, Tugnoli C, Rigillo A, Bacci B, Roccabianca P. Tumor-infiltrating lymphocytes vary in different canine soft tissue sarcoma histological types. Vet Pathol 2025; 62:276-283. [PMID: 39651618 DOI: 10.1177/03009858241300556] [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: 12/11/2024]
Abstract
Soft tissue sarcomas (STSs) are conventionally viewed as poorly immunogenic tumors; however, some human STSs have recently been reported to elicit an immune response, thus representing potential candidates for immunotherapy. Data regarding immune cell infiltrates in canine STSs are limited and reported without tumor-type stratification. The aim of this study was to retrospectively assess tumor-infiltrating lymphocytes (TILs) in canine STSs of 5 different histotypes. Eighty-seven canine STSs were collected: 22 perivascular wall tumors (PWTs), 19 liposarcomas, 17 fibrosarcomas, 16 myxosarcomas, and 13 leiomyosarcomas. The tumors were graded and immunolabeled for CD3, CD20, and FoxP3, and slides were scanned. T-cell, B-cell, Treg, and total TIL densities were quantified with QuPath software and expressed as cells/mm2. The B/T-cells ratio and Treg/T-cell proportions were calculated. Total TIL densities were higher in PWTs and myxosarcomas (median = 225 and 303, respectively). PWTs had higher T-cell density but lower Treg proportion (median = 152 and 7.6% respectively). Myxosarcomas had higher Treg densities and B/T-cell ratios (median = 24.4 and 1.57, respectively). No association with grade was found among STSs as a group. In myxosarcomas, higher grade was significantly associated with higher total TILs, and CD20+ and FoxP3+ cell densities (p < .05). The results suggest that PWTs and myxosarcomas may represent the most immunogenic STS types. Myxosarcomas elicit a B-cell and Treg-rich immune response; PWTs stimulate a T-cell-rich and Treg-poor reaction. The immune system response may contribute to the more aggressive behavior of myxosarcomas and the more indolent course of PWTs.
Collapse
|
29
|
Torres MB, Leung CH, Zoghbi M, Lazcano R, Ingram D, Wani K, Keung EZ, Zarzour MA, Scally CP, Hunt KK, Conley A, Bishop AJ, Guadagnolo BA, Farooqi A, Mitra D, Yoder AK, Nakazawa MS, Araujo D, Livingston A, Ratan R, Patel S, Ravi V, Lazar AJ, Roland CL, Somaiah N, Nassif Haddad EF. Dedifferentiated liposarcomas treated with immune checkpoint blockade: the MD Anderson experience. Front Immunol 2025; 16:1567736. [PMID: 40370451 PMCID: PMC12075363 DOI: 10.3389/fimmu.2025.1567736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/11/2025] [Indexed: 05/16/2025] Open
Abstract
Background Dedifferentiated liposarcoma (DDLPS) is one of the most common types of soft tissue sarcoma (STS) characterized by liposarcomatous differentiation and a predilection for the retroperitoneum. Despite the growing number of histology-specific immune checkpoint blockade (ICB) trials in STS, it is still difficult to identify the radiographic objective response rate (ORR) for DDLPS in the real world setting. This study aimed to evaluate the ORR and survival of patients with DDLPS treated with ICB at a single center. Methods We conducted a retrospective study of 31 patients with pathologically confirmed DDLPS treated with ICB at MD Anderson Cancer Center between 2018 and 2023. Patient demographics, disease characteristics, treatment history, and response to ICB were analyzed. Immunohistochemical analysis was performed on tumor samples to assess immune-related markers. Results ORR by RECIST 1.1 was 3.2% (n=1/31). Among all patients (n=31), 6% achieved partial radiographic response, while 39% had stable disease, and 55% showed progressive disease. Median progression-free survival (PFS) was 3.5 (95%CI:1.9, 4.7) months, and overall survival (OS) after ICB initiation was 19.7 (95%CI: 8.8, not reached) months. Patients without prior systemic therapy demonstrated better OS (p=0.004). Immunohistochemistry revealed no relationship between pre- or post-ICB expression of CD8, CD20, CD21 and PDL-1 and response. Conclusion While the response to ICB in DDLPS remains limited, specific immune markers may influence treatment outcomes. CD20/21 post-ICB appear more important for prognosis. Further research is warranted to identify predictive factors for ICB efficacy in DDLPS.
Collapse
Affiliation(s)
- Madeline B. Torres
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Surgery, Cooper University Hospital, Cooper Medical School of Rowan University, Camden, NJ, United States
| | - Cheuk Hong Leung
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marianne Zoghbi
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rossana Lazcano
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Davis Ingram
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Khalida Wani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Emily Z. Keung
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - M. Alejandra Zarzour
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Christopher P. Scally
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kelly K. Hunt
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anthony Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrew J. Bishop
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - B. Ashleigh Guadagnolo
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ahsan Farooqi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Devarati Mitra
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alison K. Yoder
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael S. Nakazawa
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dejka Araujo
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrew Livingston
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ravin Ratan
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shreyaskumar Patel
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vinod Ravi
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alexander J. Lazar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Christina L. Roland
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elise F. Nassif Haddad
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
30
|
Yang JI, Moresco P, Fearon D, Yao M. Identification of B cell antigens in solid cancer: initial insights and functional implications. Front Immunol 2025; 16:1571570. [PMID: 40356924 PMCID: PMC12066463 DOI: 10.3389/fimmu.2025.1571570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/01/2025] [Indexed: 05/15/2025] Open
Abstract
Cancer antigen discovery has mostly focused on T cell antigens, while antigens driving B cell responses have been largely overlooked despite representing another important branch of adaptive immune responses in cancer. Traditional B cell antigens in cancer have been studied using serological approaches analyzing polyclonal antibodies in serum. With recent technological advances in single-cell sequencing, a few studies have begun to investigate single B cell antigen specificity in the tumor microenvironment using immunoglobulin single-cell sequencing, recombinant monoclonal antibody production, cancer binding screening, and antigen identification. In this review, we highlight the initial insights into B cell directed cancer antigens and categorize them into cancer-associated viral antigens and non-viral antigens, with the latter featuring autoantigens. We will further discuss the functions of B cells in cancer in the context of their antigen specificity, and categorize their functions into antibody effector function, T cell activation, and B cell secretion. Lastly, we will provide perspectives on the challenges and opportunities in the identification of new B cell cancer antigens and highlight their translational potential.
Collapse
Affiliation(s)
- Jung-In Yang
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| | - Philip Moresco
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, United States
- Medical Scientist Training Program, Stony Brook University Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Douglas Fearon
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| | - Min Yao
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, NY, United States
| |
Collapse
|
31
|
Medina-Ceballos E, Giner F, Machado I, Heras-Morán B, Espino M, Navarro S, Llombart-Bosch A. The Prognostic Impact of the Tumor Immune Microenvironment in Synovial Sarcoma: An Immunohistochemical Analysis Using Digital Pathology and Conventional Interpretation. J Pers Med 2025; 15:169. [PMID: 40423041 DOI: 10.3390/jpm15050169] [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: 02/25/2025] [Revised: 04/17/2025] [Accepted: 04/23/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Innate and adaptive immune responses serve a crucial role in neoplasms. The interaction of immune cells with the neoplastic tissue influences tumor behavior, resulting in either pro-tumorigenic or anti-tumorigenic effects. However, the prognostic significance of the tumor immune microenvironment (TIME) in synovial sarcoma (SS) remains poorly studied. This study aimed to analyze the TIME of SS to determine its impact on the prognosis by examining the intratumoral lymphocytic and macrophagic infiltrate and its potential correlation with survival and recurrence. METHODS We conducted a retrospective observational study of 49 fusion-confirmed SS cases collected from two different institutions. We obtained clinical and follow-up data, and SSs were histologically classified according to WHO criteria. Immunohistochemical analysis, including of CD163, CD68, CD3, CD8, and CD20, was conducted in tissue microarrays using an analog scale. We examined the whole-slide tissue for the 23 cases with sufficient material available and then assessed the positive area by scanning the slides and analyzing the images using QuPath (0.4.4, Belfast, Northern Ireland) to calculate the positive area in an immune hotspot. We correlated the expression of these markers with clinical outcomes. A log-rank test and Kaplan-Meyer curves were used as appropriate (significance: p ≤ 0.05). RESULTS The most frequent morphological subtype was monophasic (59.6%), followed by biphasic (26.9%) and undifferentiated (7%). The mean disease specific survival (DSS) was 55.3 months, with a median of 33 months. The median overall survival (OS) was 50 months (range: 2-336 months). Both evaluation methods showed a good correlation for all antibodies, with Chi-square values of p < 0.05. All cases showed variable amounts of CD163-positive macrophages. The cases that showed a higher density of CD163-positive macrophages in whole-slide images subjected to digital analysis demonstrated an improved OS and DSS on Kaplan-Meier curves. Cases with lower CD8 and CD3 positivity showed a tendency toward faster progression and a slightly worse prognosis. CONCLUSIONS The tumor immune microenvironment in sarcomas is a complex system that requires further investigation to fully understand its impact on tumorigenesis and patient clinical outcomes. Our results demonstrate that a higher amount of intratumoral CD163-positive macrophage infiltrate is associated with an increased OS and DSS. Our findings show that digital pathology is more precise than subjective quantitative analysis.
Collapse
Affiliation(s)
| | - Francisco Giner
- Pathology Department, University of Valencia, 46010 Valencia, Spain
- Pathology Department, University Hospital La Fe, 46010 Valencia, Spain
| | - Isidro Machado
- Pathology Department, University of Valencia, 46010 Valencia, Spain
- Pathology Department, Instituto Valenciano de Oncología, 46009 Valencia, Spain
- Patologika Laboratory, Quirón-Salud, 46010 Valencia, Spain
- Cancer CIBER (CIBERONC), 28029 Madrid, Spain
| | - Begoña Heras-Morán
- Pathology Department, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Mónica Espino
- Pathology Department, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Samuel Navarro
- Pathology Department, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
- Pathology Department, University of Valencia, 46010 Valencia, Spain
- Cancer CIBER (CIBERONC), 28029 Madrid, Spain
| | | |
Collapse
|
32
|
Martin-Broto J, Moura DS, Hindi N. Which sarcoma requires PD1/PDL1 inhibitors, and what should be the best scheme? Present status and next steps. Curr Opin Oncol 2025:00001622-990000000-00254. [PMID: 40421971 DOI: 10.1097/cco.0000000000001149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
PURPOSE OF REVIEW The introduction of immune checkpoint inhibitors (ICI) in advanced sarcoma has been largely disappointing due to their "cold" tumor microenvironment, characterized by low tumor mutational burden, scarce CD8+ T-cell infiltration, and minimal expression of PD-1/PD-L1. However, recent findings highlight several scenarios in which immune checkpoint blockade exhibits clinical efficacy. RECENT FINDINGS ICIs have shown durable efficacy in specific sarcoma subtypes, such as alveolar soft part sarcoma (ASPS), with objective response rates (ORR) exceeding 35% and 50%, in monotherapy or in combination, respectively. Doxorubicin-based regimens plus ICIs have yielded notorious and higher ORRs in the most common sarcoma subtypes, than historical chemotherapy data. Neoadjuvant radiation therapy combined with ICIs has significantly improved disease-free survival in localized selected soft tissue sarcomas. SUMMARY Immunotherapy targeting immune checkpoints in sarcomas is evolving, with recent findings highlighting its potential. Single-arm trials underscore the efficacy of ICIs in rare sarcomas, exemplified by the FDA approval of atezolizumab for ASPS. Combination strategies are proving more effective than chemotherapy alone, with ongoing comparative studies assessing chemo-immunotherapy in both metastatic and localized sarcomas. Advances in predictive biomarkers could expand the clinical use of ICIs.
Collapse
Affiliation(s)
- Javier Martin-Broto
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital; University Hospital General de Villalba, and Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM)
| | - David S Moura
- Research Health Institute of Fundacion Jimenez Diaz (IIS/FJD; UAM) and University Hospital General de Villalba, Madrid, Spain
| | - Nadia Hindi
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital; University Hospital General de Villalba, and Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD; UAM)
| |
Collapse
|
33
|
Aobo Z, Xiao Z, Chengfei X, Zhe X, Yingxue C, Chenhe Z, Fuan X, Fan Y, Mengmeng X, Feng Y, Wengang L. Combination of immune checkpoint inhibitors and anthracyclines as a potential first-line regimen for dedifferentiated liposarcoma: systematic review and meta-analysis. Cancer Immunol Immunother 2025; 74:179. [PMID: 40257618 PMCID: PMC12011665 DOI: 10.1007/s00262-025-04007-1] [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: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION Dedifferentiated liposarcoma (DDLPS) is a rare and aggressive subtype of soft tissue sarcoma, characterized by limited treatment options and poor prognosis. Despite surgical resection being the only potentially curative treatment for localized DDLPS, the recurrence rate remains high, and systemic chemotherapy, typically anthracycline-based, shows limited efficacy in advanced stages. While immune checkpoint inhibitors (ICIs) have shown promise in various sarcoma subtypes, including DDLPS, their role as a first-line treatment remains unclear. METHODS We conducted a systematic meta-analysis to evaluate the efficacy of ICIs in treating patients with DDLPS. A total of 25 studies encompassing 245 patients were included. Data on overall response rate (ORR), progression-free survival, and grade III-V treatment-related adverse events were analyzed. We assessed treatment efficacy based on the line of therapy and treatment regimens, including ICI monotherapy, dual ICI therapy, and ICI combinations with other modalities. RESULTS The pooled ORR for all ICI-based treatments was 7%. First-line ICI therapy yielded a significantly higher ORR of 22%, compared to 4% in later-line treatment. The combination of ICI with anthracyclines demonstrated the highest ORR of 52%. In contrast, ICI regimens combined with trabectedin or other agents showed limited efficacy. Sensitivity analysis confirmed the stability of results, and publication bias was not detected. CONCLUSION This meta-analysis supports the potential role of ICIs, particularly in combination with anthracyclines, as a first-line therapeutic strategy for DDLPS. These results provide a foundation for future prospective studies aimed at optimizing immunotherapy approaches for this rare and challenging malignancy.
Collapse
Affiliation(s)
- Zhuang Aobo
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhou Xiao
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xu Chengfei
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Xi Zhe
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Chen Yingxue
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhang Chenhe
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xie Fuan
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yang Fan
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao Mengmeng
- Department of Retroperitoneal Tumor Surgery, Peking University People's Hospital, Beijing, China.
| | - Ye Feng
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Li Wengang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
34
|
Fan Z, Yi Z, Li S, He J. Parabacteroides distasonis promotes CXCL9 secretion of tumor-associated macrophages and enhances CD8 +T cell activity to trigger anti-tumor immunity against anti-PD-1 treatment in non-small cell lung cancer mice. BMC Biotechnol 2025; 25:30. [PMID: 40241108 PMCID: PMC12004837 DOI: 10.1186/s12896-025-00963-9] [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: 09/26/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Parabacteroides distasonis (P. distasonis) could regulate inflammatory markers, promote intestinal barrier integrity, and block tumor formation in colon. However, the regulatory effect of P. distasonis on non-small cell lung cancer (NSCLC) remains unknown. This study aimed to investigate the regulatory effect of P. distasonis on NSCLC and its impact on tumor immunity. METHODS We first established a mouse model of Lewis lung cancer, and administered P. distasonis and intrabitoneal injection of anti-mouse PD-1 monoclonal antibody to assess the impact of P. distasonis on tumor immunity, and mouse intestinal barrier. Then, we explored the effect of P. distasonis on CD8+T cells and CXCL9 secretion mediated by tumor-associated macrophages (TAM). We used the TLR1/2 complex inhibitor CPT22 to evaluate its effect on macrophage activation. Finally, we explored the effect of P. distasonis on CD8+T cells and CXCL9 secreted by TAM in vivo. RESULTS In vivo, P. distasonis enhanced anti-tumor effects of anti-PD-1 in NSCLC mice, improved intestinal barrier integrity, recruited macrophages, and promoted M1 polarization. In vitro, CD86 and iNOS levels in BMDM were elevated and CD206 and Arg1 levels were suppressed in membrane fraction of P. distasonis (PdMb) group in comparison to Control group. With additional CPT22 pre-treatment, the levels of CD86 and iNOS in BMDM were reduced, and the levels of CD206 and Arg1 were increased. Compared to PBS group, P. distasonis group exhibited higher proportion of CD8+T cells in tumor tissues, along with increased positive proportion of GZMB and IFN-γ in CD8+T cells. Additionally, in comparison to Control group, PdMb group showed an elevated proportion of GZMB+T and IFN-γ+T cells within CD8+T cells, and secretion of IFN-γ, TNF-α, perforin, and GZMB in CD8+T cell supernatant increased. Moreover, the proportion of CXCL9+F4/80+ macrophages in tumor tissues was higher in P. distasonis group compared to PBS group. In comparison to Control group, CXCL9 protein level in BMDM and CXCL9 secretion level in BMDM supernatant were increased in PdMb group. Finally, P. distasonis enhanced CD8+T cell activity by secreting CXCL9 from macrophages in vivo. CONCLUSIONS P. distasonis promoted CXCL9 secretion of TAM and enhanced CD8+T cell activity to trigger anti-tumor immunity against anti-PD-1 treatment in NSCLC mice.
Collapse
Affiliation(s)
- Zhijun Fan
- Department of Cardiothoracic Surgery, The People's Hospital of Liuyang, Changsha, China
| | - Zheng Yi
- Department of Cardiothoracic Surgery, The People's Hospital of Liuyang, Changsha, China
| | - Sheng Li
- Department of Gastrointestinal Surgery, The Central Hospital of Shaoyang, Shaoyang, China
| | - Junjun He
- Department of Gastrointestinal Surgery, The Central Hospital of Shaoyang, Shaoyang, China.
| |
Collapse
|
35
|
Yu J, Fu L, Wu R, Che L, Liu G, Ran Q, Xia Z, Liang X, Zhao G. Immunocytes in the tumor microenvironment: recent updates and interconnections. Front Immunol 2025; 16:1517959. [PMID: 40297580 PMCID: PMC12034658 DOI: 10.3389/fimmu.2025.1517959] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
The tumor microenvironment (TME) is a complex, dynamic ecosystem where tumor cells interact with diverse immune and stromal cell types. This review provides an overview of the TME's evolving composition, emphasizing its transition from an early pro-inflammatory, immune-promoting state to a later immunosuppressive milieu characterized by metabolic reprogramming and hypoxia. It highlights the dual roles of key immunocytes-including T lymphocytes, natural killer cells, macrophages, dendritic cells, and myeloid-derived suppressor cells-which can either inhibit or support tumor progression based on their phenotypic polarization and local metabolic conditions. The article further elucidates mechanisms of immune cell plasticity, such as the M1/M2 macrophage switch and the balance between effector T cells and regulatory T cells, underscoring their impact on tumor growth and metastasis. Additionally, emerging therapeutic strategies, including checkpoint inhibitors and chimeric antigen receptor (CAR) T and NK cell therapies, as well as approaches targeting metabolic pathways, are discussed as promising avenues to reinvigorate antitumor immunity. By integrating recent molecular insights and clinical advancements, the review underscores the importance of deciphering the interplay between immunocytes and the TME to develop more effective cancer immunotherapies.
Collapse
Affiliation(s)
- Jiyao Yu
- Department of Ultrasound, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Li Fu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Rui Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurosurgery, Jiangyou People’s Hospital, Mianyang, China
| | - Linyi Che
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qinwen Ran
- General Practice Department, Wufu Town Hospital, Chongqing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China
| | - Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
36
|
Braden J, Potter A, Rawson RV, Adegoke NA, Lo SN, Conway JW, Menzies AM, Carlino MS, Au-Yeung G, Saw RPM, Spillane AJ, Shannon KF, Pennington TE, Ch'ng S, Gyorki DE, Howle JR, Wilmott JS, Scolyer RA, Long GV, Pires da Silva I. Longitudinal Analysis Reveals Dynamic Changes in Histopathologic Features in Responders to Neoadjuvant Treatment in a Stage III BRAF-Mutant Melanoma Cohort. Mod Pathol 2025; 38:100776. [PMID: 40239808 DOI: 10.1016/j.modpat.2025.100776] [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: 12/12/2024] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Despite advances in systemic therapies, cutaneous melanoma remains a highly deadly disease. Patients with high-risk stage III melanoma have a significant likelihood of recurrence following surgery. Although adjuvant immunotherapy has been the standard of care, recent evidence demonstrates that neoadjuvant immunotherapy is more effective for higher-risk stage III patients, showing superior survival outcomes compared with adjuvant immunotherapy. This has led to an immediate paradigm shift in clinical practice toward neoadjuvant therapy for this cohort. The NeoTrio clinical trial assessed the efficacy of sequential or combination BRAF-targeted therapy with anti-programmed cell death-1 in the neoadjuvant setting. However, research on longitudinal histopathologic changes during this treatment period remains limited. Analysis of hematoxylin and eosin slides from 60 patients across 4 matched neoadjuvant timepoints revealed dynamic changes in a number of treatment response features. Females achieved significantly higher rates of major pathologic response (P = .002) and displayed higher levels of inflammatory fibrosis (P = .04) and hyalinized fibrosis (P = .01). The presence of tertiary lymphoid structures (P = .013) and plasma cells (P = .02) at resection was significantly associated with response. Combination scoring of histopathologic features (composite score and the immune-related pathologic response [irPR] score) was significantly associated with response early during the neoadjuvant period (composite score at week 2 on-treatment, P = .03; high irPR score at week 2 on-treatment, P = .01). A high irPR score at week 2 on-treatment was also found to be significantly associated with a lower chance of recurrence at this early neoadjuvant timepoint (P = .02). Other features associated with a lower likelihood of recurrence included increased hyalinized fibrosis (P = .015) and the presence of extensive lymphocyte density score (P = .01), tertiary lymphoid structures (P = .03), and plasma cells (P = .01). This study deepens our understanding of treatment response markers and their dynamic changes during neoadjuvant therapy. It underscores the significance of these features, particularly given their early emergence and strong associations with response and recurrence.
Collapse
Affiliation(s)
- Jorja Braden
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Alison Potter
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; NSW Health Pathology, Sydney, Australia
| | - Robert V Rawson
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia; NSW Health Pathology, Sydney, Australia; Department of Anatomical Pathology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Nurudeen A Adegoke
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Serigne N Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Jordan W Conway
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia; Department of Oncology, The Mater Hospital, Sydney, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Oncology, Westmead Hospital, Sydney, Australia; Department of Medical Oncology, Blacktown Hospital, Sydney, Australia
| | - George Au-Yeung
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia; Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Oncology, The Mater Hospital, Sydney, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Andrew J Spillane
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Oncology, The Mater Hospital, Sydney, Australia; Department of Surgical Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Kerwin F Shannon
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Oncology, The Mater Hospital, Sydney, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, Australia; Chris O'Brien Lifehouse, Sydney, Australia; Department of Head and Neck Surgery, Concord Repatriation Hospital, Sydney, Australia
| | - Thomas E Pennington
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Oncology, The Mater Hospital, Sydney, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Sydney Ch'ng
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Oncology, The Mater Hospital, Sydney, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, Australia; Chris O'Brien Lifehouse, Sydney, Australia; Department of Head and Neck Surgery, Concord Repatriation Hospital, Sydney, Australia
| | - David E Gyorki
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia; Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Julie R Howle
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Oncology, Westmead Hospital, Sydney, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia; NSW Health Pathology, Sydney, Australia; Department of Anatomical Pathology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia; Department of Oncology, The Mater Hospital, Sydney, Australia
| | - Ines Pires da Silva
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Blacktown Hospital, Sydney, Australia.
| |
Collapse
|
37
|
Wang SL, Chan TA. Navigating established and emerging biomarkers for immune checkpoint inhibitor therapy. Cancer Cell 2025; 43:641-664. [PMID: 40154483 DOI: 10.1016/j.ccell.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved outcomes of patients with many different cancers. These antibodies target molecules such as programmed cell death 1 (PD-1) or cytotoxic T lymphocyte associated protein 4 (CTLA-4) which normally function to limit immune activity. Treatment with ICIs reactivates T cells to destroy tumor cells in a highly specific manner, which in some patients, results in dramatic remissions and durable disease control. Over the last decade, much effort has been directed at characterizing factors that drive efficacy and resistance to ICI therapy. Food and Drug Administration (FDA)-approved biomarkers for ICI therapy have facilitated more judicious treatment of cancer patients and transformed the field of precision oncology. Yet, adaptive immunity against cancers is complex, and newer data have revealed the potential utility of other biomarkers. In this review, we discuss the utility of currently approved biomarkers and highlight how emerging biomarkers can further improve the identification of patients who benefit from ICIs.
Collapse
Affiliation(s)
- Stephen L Wang
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA.
| |
Collapse
|
38
|
Wilky BA, Schwartz GK, Gordon MS, El-Khoueiry AB, Bullock AJ, Henick B, Agulnik M, Singh A, Mahadevan D, Stebbing J, Delepine C, Chand D, Avagyan M, Wu W, Johnson B, Grossman JE, O'Day S, Trent JC, Jones RL, Tsimberidou AM. Botensilimab (Fc-enhanced anti-cytotoxic lymphocyte-association protein-4 antibody) Plus Balstilimab (anti-PD-1 antibody) in Patients With Relapsed/Refractory Metastatic Sarcomas. J Clin Oncol 2025; 43:1358-1368. [PMID: 39869830 PMCID: PMC11974637 DOI: 10.1200/jco-24-02524] [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: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/19/2024] [Indexed: 01/29/2025] Open
Abstract
PURPOSE Outcomes for patients with advanced sarcomas are poor and there is a high unmet need to develop novel therapies. The purpose of this phase I study was to define the safety and efficacy of botensilimab (BOT), an Fc-enhanced anti-cytotoxic lymphocyte-association protein-4 antibody, plus balstilimab (BAL), an anti-PD-1 antibody, in advanced sarcomas. METHODS BOT was administered intravenously (IV) at 1 mg/kg or 2 mg/kg once every 6 weeks in combination with BAL IV at 3 mg/kg once every 2 weeks for up to 2 years. The primary end point was to determine dose-limiting toxicities during the dose-escalation period. Secondary end points include objective response rate (ORR), duration of response (DOR), disease control rate, and progression-free survival (PFS) by RECIST 1.1. Exploratory end points include assessing patient biomarkers including tumor mutational burden, cytokines, and PD-L1 expression. RESULTS Overall, 64 patients with sarcoma were treated; all were evaluable for safety and 52 for efficacy. The most common treatment-related adverse event (TRAE) was diarrhea/colitis occurring in 35.9% of patients, with grade 3 in 6.3% of patients. No grade 4 or 5 TRAEs were reported. For all evaluable patients, ORR was 19.2% (95% CI, 9.6 to 32.5), and 27.8% (95% CI, 9.7 to 53.5) for evaluable patients with angiosarcoma (n = 18); 33.3% in visceral and 22.2% in cutaneous subtypes. Median PFS for evaluable patients was 4.4 months (95% CI, 2.8 to 6.1), with a 6-month PFS rate of 36% (95% CI, 22 to 50) and a median DOR of 21.7 months (95% CI, 1.9 to not reached). CONCLUSION The combination of BOT/BAL demonstrated promising efficacy and safety in a large cohort of heavily pretreated sarcoma patients. This encouraging activity warrants further investigation (ClinicalTrials.gov identifier: NCT03860272).
Collapse
Affiliation(s)
| | | | | | | | | | - Brian Henick
- Herbert Irving Comprehensive Cancer Center at Columbia University School of Medicine, New York, NY
| | - Mark Agulnik
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA
| | - Arun Singh
- University of California Los Angeles, Los Angeles, CA
| | - Daruka Mahadevan
- The University of Texas Health Sciences Center at San Antonio, San Antonio, TX
| | - Justin Stebbing
- Anglia Ruskin University, School of Life Sciences, Cambridge, United Kingdom
| | | | | | | | - Wei Wu
- Agenus Inc, Lexington, MA
| | | | | | | | - Jonathan C. Trent
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Robin L. Jones
- The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | | |
Collapse
|
39
|
Ferraro DA, Bisig B, Rotzinger DC, Pareja F, Missiaglia E, Voutsadakis I, Homicsko K, Digklia A. Case Report: Lasting complete response to pembrolizumab in mismatch repair-deficient cardiac sarcoma: a genomic characterization. Front Oncol 2025; 15:1485386. [PMID: 40248199 PMCID: PMC12003144 DOI: 10.3389/fonc.2025.1485386] [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: 08/23/2024] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Sarcomas are traditionally considered "cold" tumors with poor response to immunotherapy. However, evidence accumulating over the last years shows that immune checkpoint inhibitors (ICIs) may have a role in selected sarcoma patients according to predictive markers. Here, we report the case of a woman diagnosed with a primary cardiac undifferentiated sarcoma. Following failure of standard first line chemotherapy, high-throughput sequencing (HTS) revealed a high tumor mutational burden (TMB), pathogenic mutations in FAT1 and NOTCH2 and a microsatellite instability (MSI)-associated signature. Immunohistochemistry confirmed mismatch repair-deficiency (MMRd) and abundant CD8+ tumor-infiltrating lymphocytes (TILs), in the absence of tertiary lymphoid structures. The patient was, therefore, treated with the ICI pembrolizumab, reaching a complete response that continues to persist at last follow-up, more than seven years from initial diagnosis and nearly six years from initiation of ICI treatment. This case illustrates the importance of performing HTS in rare sarcomas given the availability of efficient therapies, such as those for tumors displaying high TMB or MMRd/MSI. In agreement with other reports, it supports the contention that MMRd/MSI status and high numbers of TILs are valuable predictive markers of response to immunotherapy in sarcomas.
Collapse
Affiliation(s)
- Daniela A. Ferraro
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
| | - Bettina Bisig
- Institute of Pathology, Department of Laboratory Medicine and Pathology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - David C. Rotzinger
- Department of Radiology, CHUV University Hospital, Lausanne, Switzerland
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Edoardo Missiaglia
- Institute of Pathology, Department of Laboratory Medicine and Pathology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Ioannis Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, ON, Canada
- Division of Clinical Sciences, Section of Internal Medicine, Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Krisztian Homicsko
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Antonia Digklia
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
40
|
Tasdogan A, Sullivan RJ, Katalinic A, Lebbe C, Whitaker D, Puig S, van de Poll-Franse LV, Massi D, Schadendorf D. Cutaneous melanoma. Nat Rev Dis Primers 2025; 11:23. [PMID: 40180935 DOI: 10.1038/s41572-025-00603-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 04/05/2025]
Abstract
Cutaneous melanoma is a common cancer in Australia and New Zealand, Europe, and North America, and its incidence is still increasing in many regions. Ultraviolet (UV) radiation exposure (for example, through excessive sunlight exposure) remains the primary risk factor for melanoma; however, public awareness campaigns have led to a marked reduction in mortality. In addition to genetic damage from UV radiation, specific genetic alterations have been linked to melanoma. The stage of the tumour at the time of diagnosis is of greater importance for melanoma prognosis than in almost any other cancer. Context-dependent genetic mutations that attenuate tumour-suppressive mechanisms or activate growth-promoting signalling pathways are crucial factors in the development of cutaneous melanoma. In addition to external factors such as UV radiation, the tumour microenvironment can contribute to melanoma progression, invasion and metastasis. Cutaneous melanoma treatment has improved considerably over the past decade with the discovery and development of immune checkpoint inhibitors and therapy targeting BRAF and MEK. Over the next decade, several priorities are likely to influence melanoma research and management, including the continued advance of precision medicine methods to identify the most suitable patients for the most effective treatment, with the aim of improving clinical outcomes.
Collapse
Affiliation(s)
- Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen & German Cancer Consortium (DKTK), Partner Site Essen, Essen, Germany.
- National Center for Tumour diseases (NCT-West), Campus Essen & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany.
| | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Alexander Katalinic
- Institute for Social Medicine and Epidemiology, University of Lübeck, Lübeck, Germany
| | - Celeste Lebbe
- Université Paris Cite, AP-HP Dermato-oncology and CIC, Cancer institute APHP.nord Paris cité, INSERM U976, Saint Louis Hospital, Paris, France
| | - Dagmar Whitaker
- Melanoma Advisory Board South Africa, Cape Town, South Africa
| | - Susana Puig
- Dermatology Department, IDIBAPS, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
- 8CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Lonneke V van de Poll-Franse
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
- Department of Medical and Clinical Psychology, CoRPS - Center of Research on Psychology in Somatic Diseases, Tilburg University, Tilburg, Netherlands
| | - Daniela Massi
- Section of Pathology, Department of Health Sciences, University of Florence, Florence, Italy
- Department of Molecular Pathobiology, New York University - College of Dentistry, New York, NY, USA
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen & German Cancer Consortium (DKTK), Partner Site Essen, Essen, Germany.
- National Center for Tumour diseases (NCT-West), Campus Essen & Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
41
|
Chen S, Pyne JM, Liu Y, Abraham Y, Wen Z, Palsgrove D, Xiao G, Truelson J, Myers L, Tillman B, Day A, Gordin E, Stankova L, Xie Y, Sher D, Bishop J, Gao J, Sumer BD. Nodal Yield From Neck Dissection Predicts the Anti-Tumor Immune Response in Head and Neck Cancers. Head Neck 2025; 47:1199-1208. [PMID: 39681999 PMCID: PMC11907675 DOI: 10.1002/hed.28006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Lymph node count (LNC) from neck dissection has been associated with undernutrition and survival in head and neck squamous cell carcinoma (HNSCC). As local components of the immune system, cervical lymph nodes may reflect anti-tumor immune status. This study investigates the relationship between decreased LNC, formation of tertiary lymphoid structures (TLS), and primary tumor infiltration by lymphocytes in undernourished patients. METHODS A matched-cohort study was conducted in a tertiary medical center, where neck dissection quality was standardized for a total of 384 subjects that were evaluated. Six head and neck cancer patients that underwent primary surgery including neck dissection with low LNC and BMI (low BMI < 23, low LNC ≤ 5.6 per neck level) were matched by stage, p16 status, and subsite to 16 patients with normal BMI and high LNC. Multiplexed immunohistochemistry was used to evaluate the tumor-infiltrating lymphocytes and the number and quality of TLS within primary tumors. Whole primary cancers underwent automated analysis and counting of leukocytes after multiplexed immunohistochemistry staining of tumor slides. A head and neck pathologist blindly scored the number and maturity of TLS. Descriptive statistics were used to analyze outcomes. RESULTS The patients with low BMI and low LNC had significantly fewer CD3 (p = 0.0136), CD8 (p = 0.0003), and CD20 (p = 0.0334) cells in their primary tumors compared to patients with normal BMI and LNC. The low BMI low LNC patients also had fewer mature TLS (0.83/tumor) in their primary cancers compared to patients with normal BMI and high LNC (5.4/tumor) and also had greater than fourfold lower mature TLS density (TLS per μm2 mean) (6.34 × 10-9 vs. 2.82 × 10-8), with significantly worsened survival relative to patients with low BMI and normal LNC and patients with normal BMI. CONCLUSION Low LNC predicts worsened survival only in low BMI HNSCC patients with non-HPV related tumors and in these patients is associated with markers of immunosuppression such as fewer tumor-infiltrating CD8+ T-cells, CD20+ cells, and fewer TLS in primary cancers compared to matched normal BMI patients with high LNC.
Collapse
Affiliation(s)
- Shuqing Chen
- Department of Otolaryngology—Head & Neck SurgeryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Justin M. Pyne
- Department of Otolaryngology—Head & Neck SurgeryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Yulun Liu
- Department of BioinformaticsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Youstina Abraham
- Department of Otolaryngology—Head & Neck SurgeryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Zhuoyu Wen
- Department of BioinformaticsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Doreen Palsgrove
- Department of PathologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Guanghua Xiao
- Department of BioinformaticsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - John Truelson
- Department of Otolaryngology—Head & Neck SurgeryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Larry Myers
- Department of Otolaryngology—Head & Neck SurgeryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Brittny Tillman
- Department of Otolaryngology—Head & Neck SurgeryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Andrew Day
- Department of Otolaryngology—Head & Neck SurgeryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Eli Gordin
- Department of Otolaryngology—Head & Neck SurgeryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Lenka Stankova
- Department of Otolaryngology—Head & Neck SurgeryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Yang Xie
- Department of BioinformaticsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - David Sher
- Department of Radiation OncologySimmons Cancer CenterDallasTexasUSA
| | - Justin Bishop
- Department of PathologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Jinming Gao
- Department of OtolayngologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Baran D. Sumer
- Department of Otolaryngology—Head & Neck SurgeryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| |
Collapse
|
42
|
Chu H, Li Y, Yang H, Liu Y, Zheng R, Zhang X, Wang X, Zhao J, Zhang Y, Wang Q, Ran Y, Guo L, Zhou S, Liu M, Song W, Wang B, Li L, Zhou L. Characterisation and Clinical Relevance of Tertiary Lymphoid Structures in Primary Biliary Cholangitis. Liver Int 2025; 45:e16157. [PMID: 39552515 DOI: 10.1111/liv.16157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 09/30/2024] [Accepted: 10/25/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND AND AIMS The pathological characteristics of lymphocyte infiltration in the hepatic portal tracts of patients with primary biliary cholangitis (PBC) remain unclear. Tertiary lymphoid structures (TLSs) are ectopic lymphoid tissues associated with the exacerbation of autoimmune reactions. Here, we evaluate the role of TLSs in PBC and investigate their potential therapeutic value. METHODS We recruited 75 patients with PBC and 53 control patients with liver biopsies who were followed more than 2 years. TLSs and their maturity were identified by the amount and spatial distribution of immune cells. Bulk RNA sequencing of liver was performed in PBC patients with different TLS maturity. The sphingosine-1-phosphate receptor (S1PRs) modulator FTY720 was administered to dnTGFβRII mice to assess the role of TLSs on cholangitis. RESULTS TLSs presented in 61.3% (46/75) of liver tissues from patients with PBC, including 26 patients with mature TLS (mTLS) and 20 patients with immature TLS (imTLS). The proportion of mTLS was higher in PBC compared with chronic hepatitis B and autoimmune hepatitis. PBC patients with mTLS exhibited the highest serum levels of biochemical indicators, immune globulin and proportions of liver cirrhosis. Gene sets for lymphocyte migration and chemokine signalling pathways were enriched in patients with PBC presenting with TLS. FTY720 inhibited TLS formation and relieved cholangitis and fibrosis in dnTGFβRII mice. CONCLUSION TLSs are characteristics of lymphocyte accumulation in the portal tracts of PBC, of which the maturity of TLSs correlates with the inflammation and fibrosis of PBC. Targeting TLSs formation is a potential treatment of PBC.
Collapse
Affiliation(s)
- Hongyu Chu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yanni Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hui Yang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yuhang Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Rongrong Zheng
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xue Zhang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xiaoyi Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jingwen Zhao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yujie Zhang
- Department of Pathology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Quan Wang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ying Ran
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Liping Guo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Simin Zhou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Man Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Wenjing Song
- Department of Pathology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Long Li
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lu Zhou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| |
Collapse
|
43
|
Schneider D, Brown EDL, Gluski J, Mishra A, Shah HA, Sciubba DM, Lo SFL. Subtype-Specific Patterns of Tumor Purity and Mutation Load Suggest Treatment Implications: A Cross-Sectional Analysis of 7494 Soft Tissue and Bone Sarcomas (MSK Cohort). Am J Clin Oncol 2025; 48:185-192. [PMID: 40085522 DOI: 10.1097/coc.0000000000001161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
OBJECTIVES Sarcomas are complex mesenchymal malignancies whose molecular characteristics can significantly influence treatment strategies. This study aimed to investigate the relationship between tumor purity, mutation load, and clinical characteristics across sarcoma subtypes, focusing on potential implications for therapeutic stratification. METHODS This study analyzed the molecular characteristics of 7494 sarcoma cases from the Soft Tissue and Bone Sarcoma (MSK, Nat Commun 2022) data set using available case analysis. Correlations between tumor purity, mutation load, age, and sex were analyzed using nonparametric methods, with subtype-specific analyses conducted using Kruskal-Wallis tests and Bonferroni-corrected post hoc comparisons. A comprehensive analysis of mutation patterns was performed using microsatellite instability (MSI) status. RESULTS Significant correlations between mutation load and tumor purity (ρ=0.320, P <0.001) were identified, with marked heterogeneity across subtypes. Tumor purity ranged from 20.0% in brain sarcomas to 78.5% in dermatofibrosarcoma protuberans. Age-related molecular changes were observed in brain (ρ=0.711, P =0.006) and skin sarcomas (ρ=0.450, P =0.006), suggesting distinct evolutionary patterns. A subset of hypermutated, microsatellite stable cases (0.15%) with mutation loads exceeding 100 mutations/mb were identified, suggesting alternative mechanisms of genomic instability. MSI-high status was rare (0.24%) but associated with higher mutation loads (median: 25.84 vs. 2.42, P <0.001), particularly in uterine sarcomas (0.7% prevalence). CONCLUSIONS The identification of distinct molecular patterns across sarcoma subtypes challenge existing morphology-based classification systems and may hold implications for therapeutic stratification. These findings may help inform future immunotherapeutic and molecular-guided approaches to treatment in sarcoma patients, particularly for elderly patients with brain sarcomas or females with uterine sarcomas.
Collapse
Affiliation(s)
- Daniel Schneider
- Department of Neurosurgery, Donald and Barbara Zucker Hofstra School of Medicine at Northwell, Manhasset, NY
| | | | | | | | | | | | | |
Collapse
|
44
|
Cui X, Gu X, Li D, Wu P, Sun N, Zhang C, He J. Tertiary lymphoid structures as a biomarker in immunotherapy and beyond: Advancing towards clinical application. Cancer Lett 2025; 613:217491. [PMID: 39862919 DOI: 10.1016/j.canlet.2025.217491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Tertiary lymphoid structures (TLSs) are ectopic immune cell clusters formed in nonlymphoid tissues affected by persistent inflammation, such as in cancer and prolonged infections. They have features of the structure and function of secondary lymphoid organs, featuring central CD20+ B cells, surrounded by CD3+ T cells, CD21+ follicular dendritic cells, and CD68+ macrophages, with a complex vascular system. TLS formation is governed by lymphotoxin-α1β2, TNF, and chemokines like CCL19, CCL21, and CXCL13, differing from secondary lymphoid organ development in developing later in life at sites of chronic inflammation. Their role in enhancing immune responses, particularly in the context of cancer, makes them a focal point in immunotherapy. This review discusses recent advances in TLS assessment that involves complex gene expression signatures, histological analysis, artificial intelligence, and spatial omics. The presence and maturity of TLS are associated with better outcomes in various cancers, acting as a biomarker for immunotherapy effectiveness. This review explores the structure, formation, and role of TLS in disease prognosis, including their roles in immunotherapy and non-immunotherapy treatments, highlighting a need to develop novel techniques for precise characterization of TLS as well as their significance as predictive biomarkers beyond traditional biomarkers.
Collapse
Affiliation(s)
- Xinyu Cui
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Xuanyu Gu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Dongyu Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Peng Wu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
45
|
Wu Y, Zhu L, Li S, Liu L, Wang Y, Yang Y, Mu Y, Zhu Q, Jiang Y, Wu C, Xi P, Ma C, Liang L, Gao M, Hu Y, Ding Q, Pan S. DA-DRD5 signaling reprograms B cells to promote CD8 + T cell-mediated antitumor immunity. Cell Rep 2025; 44:115364. [PMID: 40023842 DOI: 10.1016/j.celrep.2025.115364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/16/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025] Open
Abstract
Neuronal signals have emerged as pivotal regulators of B cells that regulate antitumor immunity and tumor progression. However, the functional relevance and mechanistic basis of the effects of the neurotransmitter dopamine (DA) on tumor immunity remain elusive. Here, we discovered that plasma DA levels are positively correlated with circulating B cell numbers and potently activate B cell responses in a manner dependent on the DRD5 receptor. Notably, DRD5 signaling enhanced the Janus kinase 1 (JAK1)-STAT1 signaling in B cell responses, which enhanced B cell activation and increased antigen presentation and co-stimulation, resulting in increased expansion and cytotoxicity in tumor-specific effector of T cells. Our findings demonstrate that DA signaling suppresses tumor progression and highlight DRD5 as a promising target for cancer immunotherapy.
Collapse
Affiliation(s)
- Yuqing Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Lei Zhu
- Jiangsu Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210036, China; Department of Breast Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing 210004, China
| | - Sheng Li
- Department of Clinical Laboratory, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Lu Liu
- Department of Immunology, Nanjing Medical University, Nanjing 211166, China
| | - Yaman Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Yongbing Yang
- Department of Medical Laboratory, Affiliated Children's Hospital of Jiangnan University, Wuxi 214000, China
| | - Yuan Mu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Qiuying Zhu
- The First Clinical School of Nanjing Medical University, Nanjing 210029, China
| | - Yuying Jiang
- Department of Immunology, Nanjing Medical University, Nanjing 211166, China
| | - Chunyan Wu
- Department of Pathology, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing 210004, China
| | - Peiwen Xi
- Department of Health Management Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chunmei Ma
- Department of Immunology, Nanjing Medical University, Nanjing 211166, China
| | - Lijun Liang
- Department of Thoracic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Min Gao
- Department of Nephrology, Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200336, China
| | - Yingchao Hu
- Department of Immunology, Nanjing Medical University, Nanjing 211166, China.
| | - Qiang Ding
- Jiangsu Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210036, China.
| | - Shiyang Pan
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China.
| |
Collapse
|
46
|
Zhang H, Zhang Y, Zhou P, Guo Y, Jiang L, Gu J. Intra-tumoural RAMP1+ B cells promote resistance to neoadjuvant anti-PD-1-based therapy in oesophageal squamous cell carcinoma. IMMUNOTHERAPY ADVANCES 2025; 5:ltaf012. [PMID: 40385640 PMCID: PMC12084762 DOI: 10.1093/immadv/ltaf012] [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] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 03/17/2025] [Indexed: 05/20/2025] Open
Abstract
Introduction The application of neoadjuvant immunotherapy in oesophageal squamous cell carcinoma (ESCC) reactivates anti-tumour immune responses and prolong postoperative survival. However, due to the heterogeneity of tumour microenvironment, limited patients have achieved pathological regression after treatment. The dual roles of B cells were recently highlighted in ESCC. The study aimed to investigate the role of B cell subclusters and the upstream signalling of B cell differentiation in ESCC resistant to immunotherapy. Methods Single-cell RNA sequencing was employed for ESCC specimens with distinct responses to neoadjuvant immunotherapy to map the landscape of intra-tumoural B cells. Results A novel subset of neuropeptide receptor, receptor activity-modifying protein 1 (RAMP1) positive B cells was revealed to accumulate in ESCC that is resistant to neoadjuvant immunotherapy. Stimulated by nociceptor neurons secreting calcitonin gene-related peptide (CGRP), RAMP1(+) B cells exhibit an immunosuppressive phenotype. The elevated secretion of immune-regulating cytokines by RAMP1(+) B cells blunts the cytotoxicity of Cluster of Differentiation (CD)8(+) T cell and leads to tumour immune evasion. A combination of RAMP1 blocker and anti-Programmed cell death protein (PD)-1 therapies synergistically reinvigorated anti-tumour immunity, reducing tumour progression in vitro. Conclusion The study suggests that RAMP1(+) B cells play a critical role in mediating resistance to neoadjuvant immunotherapy in ESCC. Targeting the CGRP-RAMP axis remodels B cells and enhance the efficacy of current immunotherapies, providing new strategies for overcoming treatment resistance.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuchen Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pingjing Zhou
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifan Guo
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liqun Jiang
- Department of Thoracic Surgery, Jingjiang People’s Hospital Affiliated to Yangzhou University, Jingjiang, China
| | - Jie Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Gao T, He X, Wang J, Liu J, Hu X, Bai C, Yin S, Shi Y, Wang Y, Tan Z, Cao F, Li S, Shi YJ, Xue R, Li J, He Y, Li J, Lu H, Zhang H, Zhang L, Fang Z, Wang X, Liu M, Fu W, Tang L, Ye B, Fan Z, Xi JJ. Self-assembled patient-derived tumor-like cell clusters for personalized drug testing in diverse sarcomas. Cell Rep Med 2025; 6:101990. [PMID: 40054460 PMCID: PMC11970405 DOI: 10.1016/j.xcrm.2025.101990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/30/2024] [Accepted: 02/04/2025] [Indexed: 03/21/2025]
Abstract
Several patient-derived tumor models have emerged recently. However, soft tissue sarcomas (STSs) present a challenge in developing preclinical drug-testing models due to their non-epithelial and complex nature. Here, we report a model termed patient-derived tumor-like cell clusters (PTCs) derived from STS patients. PTCs result from the self-assembly and proliferation of mesenchymal stem cells (MSCs), epithelial cells, and immune cells, faithfully recapitulating the morphology and function of the original tumors. Through standardized culture and drug-response assessment protocols, PTCs facilitate personalized drug testing, evaluating hundreds of therapies within two weeks. Notably, PTCs exhibit 100% accuracy in distinguishing between complete or partial response and disease progression. We demonstrate the utility of PTCs in guiding chemotherapy selection for a patient with relapse and metastases following conventional therapy, who exhibited a positive response after non-conventional therapy identified through PTC. These findings underscore the potential of PTCs for prospective use in clinical decision-making regarding therapy selection.
Collapse
Affiliation(s)
- Tian Gao
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xinyu He
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Junyi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jiayong Liu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiongbing Hu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Chujie Bai
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shenyi Yin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; GeneX Health Co., Ltd., Beijing 100195, China
| | - Yunfei Shi
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yanmin Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Zhichao Tan
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fang Cao
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shu Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yan-Jie Shi
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ruifeng Xue
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Juan Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Yang He
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jiaxin Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China
| | - Huinan Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China
| | - Hanshuo Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; GeneX Health Co., Ltd., Beijing 100195, China
| | - Lu Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhiwei Fang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xinyu Wang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Mengmeng Liu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wenjun Fu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lei Tang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Buqing Ye
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Zhengfu Fan
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Jianzhong Jeff Xi
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China.
| |
Collapse
|
48
|
Jing SY, Wang HQ, Lin P, Yuan J, Tang ZX, Li H. Quantifying and interpreting biologically meaningful spatial signatures within tumor microenvironments. NPJ Precis Oncol 2025; 9:68. [PMID: 40069556 PMCID: PMC11897387 DOI: 10.1038/s41698-025-00857-1] [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] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in orchestrating tumor cell behavior and cancer progression. Recent advances in spatial profiling technologies have uncovered novel spatial signatures, including univariate distribution patterns, bivariate spatial relationships, and higher-order structures. These signatures have the potential to revolutionize tumor mechanism and treatment. In this review, we summarize the current state of spatial signature research, highlighting computational methods to uncover spatially relevant biological significance. We discuss the impact of these advances on fundamental cancer biology and translational research, address current challenges and future research directions.
Collapse
Affiliation(s)
- Si-Yu Jing
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - He-Qi Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Ping Lin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Jiao Yuan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Zhi-Xuan Tang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Hong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China.
| |
Collapse
|
49
|
Li C, Wang P, Dong Z, Cao W, Su Y, Zhang J, Zhao S, Wang Z, Lei Z, Shi L, Cheng R, Liu W. Single-cell transcriptomics analysis reveals that the tumor-infiltrating B cells determine the indolent fate of papillary thyroid carcinoma. J Exp Clin Cancer Res 2025; 44:91. [PMID: 40069827 PMCID: PMC11895268 DOI: 10.1186/s13046-025-03341-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
OBJECTIVE Active surveillance (AS) offers a viable alternative to surgical intervention for the management of indolent papillary thyroid carcinoma (PTC), helping to minimize the incidence of unnecessary treatment. However, the broader adoption of AS is hindered by the need for more reliable diagnostic markers. This study aimed to identify the differences between indolent and progressive PTC and find new targets for biomarker development and therapeutic strategies. METHODS We used single-cell RNA sequencing (scRNA-seq) to analyze cellular differences in 10 early-stage PTC tumors. Findings were validated in an additional 25 tumors using cell co-culture, migration assays, immunofluorescence staining, flow cytometry, and analysis of data from The Cancer Genome Atlas (TCGA). RESULTS Tumor-infiltrating B cells (TIL-B), particularly germinal center B cells (GC-B), were more abundant in indolent PTC. These cells suppressed thyroid cell proliferation in both indolent and progressive cases, though indolent PTC had a higher capacity to recruit peripheral B cells. In indolent cases, TIL-B cells showed increased proliferation and formed clusters within tertiary lymphoid structures (TLS). PTPRC-CD22 interactions were identified as potential drivers of TIL-B cell proliferation. Markers linked to GC-B cells, such as LMO2, were highlighted as potential diagnostic and prognostic indicators for indolent PTC. CONCLUSION This study provides insights into the cellular landscape of early-stage PTC, revealing distinct tumor and immune microenvironment features in indolent and progressive cases. These findings advance the understanding of indolent PTC biology and support the development of reliable diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Chunmei Li
- State Key Laboratory for Conservation and Utilization of Bio-resources and School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China
| | - Pei Wang
- Department of Radiation Oncology, Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Zhizhong Dong
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Weihan Cao
- Department of Ultrasound Imaging, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yanjun Su
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jianming Zhang
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Shuyan Zhao
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhiyuan Wang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zi Lei
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Li Shi
- Endocrine and Metabolic Diseases Clinical Medical Center of Yunnan, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ruochuan Cheng
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Wen Liu
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
50
|
Zhang E, Ma Y, Liu Z, Zhang J, Liu W, Chen Y, Liu G, Liu X, Zhang F, Zhu Y, Yang Y, Tian X. Prognostic implications and characterization of tumor-associated tertiary lymphoid structures genes in pancreatic cancer. J Transl Med 2025; 23:301. [PMID: 40065365 PMCID: PMC11892293 DOI: 10.1186/s12967-025-06152-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/18/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is among the most aggressive cancers, with rising incidence and limited responsiveness to immunotherapy due to its highly suppressive tumor microenvironment (TME). Tertiary lymphoid structures (TLS), ectopic formation structures of immune cells, are linked to better prognosis and improved immunotherapy responses in PDAC. Understanding TLS's role in PDAC could enhance immunotherapy effectiveness. METHODS This study integrated transcriptomic and clinical data from 310 PDAC patients in GEO database. We performed consensus clustering using tumor-associated TLS (TA-TLS) genes, identifying three distinct molecular subtypes. Single-sample gene set enrichment analysis (ssGSEA) was then employed to calculate a TLS score for each patient, allowing for TLS-based evaluation. Key prognostic genes were identified using an iterative LASSO method, leading to the construction of a risk assessment model, which was validated across independent cohorts. We further analyzed the TLS score using single-cell RNA sequencing (scRNA-seq), visualized key gene expression, and validated protein expression through immunohistochemistry (IHC). Additionally, we explored the effects of DNASE1L3 on cell proliferation and migration, and its immune-related functions using Gene Set Enrichment Analysis (GSEA) and multiplex cytokine analysis. RESULTS Consensus clustering revealed three PDAC molecular subtypes with significant differences in prognosis, TA-TLS gene expression, and TME features. The TLS score effectively stratified patients into high and low groups, correlating with survival outcomes and TME characteristics. Our risk model, validated across cohorts, reliably predicted patient outcomes. Validation studies showed lower expression of DNASE1L3 and IL33 in tumor tissues. scRNA-seq confirmed TLS score associations with immune cells. DNASE1L3 overexpression inhibited PDAC cell proliferation and migration, with cytokine analysis indicating increased immune activity. CONCLUSIONS This study elucidated the expression profile of TA-TLS genes in PDAC, constructed a TLS gene-based scoring system, and developed a related risk model. We also explored the functions and potential antitumor mechanisms of key genes, providing evidence and new insights for enhancing TLS-targeted immunotherapy strategies in PDAC.
Collapse
Affiliation(s)
- Enkui Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yongsu Ma
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Zonghao Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Jixin Zhang
- Department of Pathology, Peking University First Hospital, Beijing, 100034, China
| | - Weikang Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yiran Chen
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Guangnian Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Xinxin Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Fusheng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yu Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yinmo Yang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China.
- Present address: No. 8 Xishiku Street, Xicheng District, Beijing, China.
| | - Xiaodong Tian
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, No. 8 Xishiku Street, Xicheng District, Beijing, 100034, China.
- Present address: No. 8 Xishiku Street, Xicheng District, Beijing, China.
| |
Collapse
|