1
|
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
|
2
|
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] [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
|
3
|
Wang X, Jiang Y, Yang S, Wang F, Zhang X, Wang W, Chen Y, Wu X, Xiang J, Li Y, Jiang X, Yuan W, Zhang J, Yu KH, Ward RL, Hawkins N, Jonnagaddala J, Li G, Li R. Foundation Model for Predicting Prognosis and Adjuvant Therapy Benefit From Digital Pathology in GI Cancers. J Clin Oncol 2025:JCO2401501. [PMID: 40168636 DOI: 10.1200/jco-24-01501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/21/2024] [Accepted: 02/06/2025] [Indexed: 04/03/2025] Open
Abstract
PURPOSE Artificial intelligence (AI) holds significant promise for improving cancer diagnosis and treatment. Here, we present a foundation AI model for prognosis prediction on the basis of standard hematoxylin and eosin-stained histopathology slides. METHODS In this multinational cohort study, we developed AI models to predict prognosis from histopathology images of patients with GI cancers. First, we trained a foundation model using over 130 million patches from 104,876 whole-slide images on the basis of self-supervised learning. Second, we fine-tuned deep learning models for predicting survival outcomes and validated them across seven cohorts, including 1,619 patients with gastric and esophageal cancers and 2,594 patients with colorectal cancer. We further assessed the model for predicting survival benefit from adjuvant chemotherapy. RESULTS The AI models predicted disease-free survival and disease-specific survival with a concordance index of 0.726-0.797 for gastric cancer and 0.714-0.757 for colorectal cancer in the validation cohorts. The models stratified patients into high-risk and low-risk groups, with 5-year survival rates of 49%-52% versus 76%-92% in gastric cancer and 43%-72% versus 81%-98% in colorectal cancer. In multivariable analysis, the AI risk scores remained an independent prognostic factor after adjusting for clinicopathologic variables. Compared with stage alone, an integrated model consisting of stage and image information improved prognosis prediction across all validation cohorts. Finally, adjuvant chemotherapy was associated with improved survival in the high-risk group but not in the low-risk group (treatment-model interaction P = .01 and .006) for stage II/III gastric and colorectal cancer, respectively. CONCLUSION The pathology foundation model can accurately predict survival outcomes and complement clinicopathologic factors in GI cancers. Pending prospective validation, it may be used to improve risk stratification and inform personalized adjuvant therapy.
Collapse
Affiliation(s)
- Xiyue Wang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Yuming Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Sen Yang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Fang Wang
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Xiaoming Zhang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Wei Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yijiang Chen
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Xiaoyan Wu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinxi Xiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Yuchen Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Xiaofeng Jiang
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Wei Yuan
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Jing Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | - Robyn L Ward
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Nicholas Hawkins
- School of Medical Sciences, University of New South Wales, Sydney, Kensington, NSW, Australia
| | | | - Guoxin Li
- School of Clinical Medicine, Tsinghua University, Beijing Tsinghua Changgung Hospital, Beijing, China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
- Stanford Institute for Human-Centered Artificial Intelligence, Stanford, CA
| |
Collapse
|
4
|
Yao Z, Fan J, Bai Y, He J, Zhang X, Zhang R, Xue L. Unravelling Cancer Immunity: Coagulation.Sig and BIRC2 as Predictive Immunotherapeutic Architects. J Cell Mol Med 2025; 29:e70525. [PMID: 40159652 PMCID: PMC11955421 DOI: 10.1111/jcmm.70525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) represent a groundbreaking advancement in cancer therapy, substantially improving patient survival rates. Our comprehensive research reveals a significant positive correlation between coagulation scores and immune-related gene expression across 30 diverse cancer types. Notably, tumours exhibiting high coagulation scores demonstrated enhanced infiltration of cytotoxic immune cells, including CD8+ T cells, natural killer (NK) cells, and macrophages. Leveraging the TCGA pan-cancer database, we developed the Coagulation.Sig model, a sophisticated predictive framework utilising a coagulation-related genes (CRGs) to forecast immunotherapy outcomes. Through rigorous analysis of ten ICI-treated cohorts, we identified and validated seven critical CRGs: BIRC2, HMGB1, STAT2, IFNAR1, BID, SPATA2, IL33 and IFNG, which form the foundation of our predictive model. Functional analyses revealed that low-risk tumours characterised by higher immune cell populations, particularly CD8+ T cells, demonstrated superior ICI responses. These tumours also exhibited increased mutation rates, elevated neoantigen loads, and greater TCR/BCR diversity. Conversely, high-risk tumours displayed pronounced intratumor heterogeneity (ITH) and elevated NRF2 pathway activity, mechanisms strongly associated with immune evasion. Experimental validation highlighted BIRC2 as a promising therapeutic target. Targeted BIRC2 knockdown, when combined with anti-PD-1 therapy, significantly suppressed tumour growth, enhanced CD8+ T cell infiltration, and amplified IFN-γ and TNF-α secretion in tumour models. Our findings position the Coagulation.Sig model as a novel, comprehensive approach to personalised cancer treatment, with BIRC2 emerging as both a predictive biomarker and a potential therapeutic intervention point.
Collapse
Affiliation(s)
- Ziang Yao
- Department of Traditional Chinese MedicinePeking University People's HospitalBeijingChina
| | - Jun Fan
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yucheng Bai
- Department of Thoracic SurgeryFirst Affiliated Hospital, Anhui Medical UniversityHefeiChina
| | - Jiakai He
- Department of Traditional Chinese MedicinePeking University People's HospitalBeijingChina
| | - Xiang Zhang
- Department of Respiratory and Critical Care MedicineThe Affiliated Huai'an Hospital of Xuzhou Medical University, the Second People's Hospital of Huai'anHuai'anJiangsuChina
| | - Renquan Zhang
- Department of Thoracic SurgeryFirst Affiliated Hospital, Anhui Medical UniversityHefeiChina
| | - Lei Xue
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
5
|
Wang H, Liu Y, Yang L, Wang Z, Hou Q, Zhang J, Huang W, Ma D, Liu Y. Differential roles of IL-17B and IL-17RB in colorectal cancer: Correlation with immune infiltration and prognosis. Pathol Res Pract 2025; 268:155847. [PMID: 40020328 DOI: 10.1016/j.prp.2025.155847] [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: 10/08/2024] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND The aim of the research is to investigate correlation of immune infiltration between IL-17B and IL-17RB in colorectal cancer (CRC), then provide an experimental basis for clinical diagnostic marker screening of CRC. METHODS Gene expression levels were assessed via TIMER and GEPIA databases, protein expression through the Human Protein Atlas (HPA), clinicopathological correlations and prognosis via UALCAN and KM-Plotter, respectively. Mutation analysis was conducted using cBioPortal, immune cell infiltration via TIMER, and hub genes were identified through protein-protein interaction (PPI) networks. Biological functions and pathways were elucidated with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Finally, the expression of IL-17B, IL-17RB, and associated inflammatory cells in CRC were analyzed using immunohistochemical staining and special staining technique. RESULTS Bioinformatics analysis showed that IL-17B gene and protein expression levels decreased, while IL-17RB expression increased in CRC. IL-17B expression was affected by gender, body weight, histology, lymph node status, and tumour grade. Overexpression of IL-17B was negatively correlated with progression-free survival in CRC. IL-17B is involved in phosphatidylinositol 3-kinase/AKT signaling, vascular development, and other processes. IL-17B is associated with mitochondrial gene expression, regulation of mRNA metabolism, amino acid metabolism and other processes, as well as phosphatidylinositol-binding and liganding. Inositol 3-kinase/AKT signalling and vascular development. IL-17B was negatively correlated with mitochondrial gene expression, regulation of mRNA metabolism, amino acid metabolism and other processes as well as with molecular functions such as phosphatidylinositol binding and ligase activity. IL-17RB expression was correlated with the clinicopathological features described above and decreased with tumour progression. High levels of IL-17RB were associated with improved overall survival and immune cell infiltration. The key genes of IL-17RB are mainly involved in DNA damage, metabolism, checkpoint signaling and regulation of replication. Immunohistochemical staining results showed that the expression of IL-17B and IL-17RB reduced in CRC, compared to normal colon tissue (p < 0.05). IL-17B was positively correlated with CD4+ T lymphocyte and mast cell infiltration. IL-17RB was positively correlated with CD4+ T lymphocyte infiltration and negatively correlated with CD20+ B lymphocyte infiltration. CONCLUSION The expression of IL-17RB in CRC decreased with increasing tumour stage, and high levels of IL-17RB predicted a better prognosis, suggesting that its decreased expression was associated with disease progression. Therefore, IL-17RB may be a biomarker for assessing the prognosis of CRC. Meanwhile, IL-17B was positively correlated with CD4+ T lymphocyte and mast cell infiltration, and its overexpression was negatively correlated with recurrence-free survival, IL-17B and IL-17RB may affect CRC through different pathway mechanisms.
Collapse
Affiliation(s)
- Han Wang
- School of Basic Medical College, Beihua University, Jilin 132013, China.
| | - Yuqi Liu
- School of Basic Medical College, Beihua University, Jilin 132013, China
| | - Lijuan Yang
- School of Basic Medical College, Beihua University, Jilin 132013, China
| | - Zhenjiang Wang
- School of Basic Medical College, Beihua University, Jilin 132013, China
| | - Qinlong Hou
- School of Basic Medical College, Beihua University, Jilin 132013, China
| | - Jihong Zhang
- The Affiliated Hospital of Beihua University, Jilin 132013, China
| | - Weili Huang
- The Affiliated Hospital of Beihua University, Jilin 132013, China
| | - Dongrui Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Yanbo Liu
- School of Basic Medical College, Beihua University, Jilin 132013, China.
| |
Collapse
|
6
|
Liao S, Zhang X, Chen L, Zhang J, Lu W, Rao M, Zhang Y, Ye Z, Ivanova D, Li F, Chen X, Wang Y, Song A, Xie B, Wang M. KRT14 is a promising prognostic biomarker of breast cancer related to immune infiltration. Mol Immunol 2025; 180:55-73. [PMID: 40014952 DOI: 10.1016/j.molimm.2025.02.016] [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: 09/13/2024] [Revised: 01/12/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Breast cancer (BC) is the leading cancer among women globally, which has the highest incidence and mortality rate in over a hundred countries. This study was intended to discover a new prognostic biomarker, facilitating personalized treatment approaches. METHODS RNA sequencing data from The Cancer Genome Atlas database and Gene Expression Omnibus database were utilized to download to evaluate expression levels and prognostic significance of Keratin 14 (KRT14). Methylation of KRT14 was also assessed. The CIBERSORT and single-sample gene set enrichment analysis algorithms were applied to explore the connection between KRT14 and the tumor microenvironment. Primary drugs' sensitivity and potential small molecule therapeutic compounds were analyzed through the "pRRophetic" R package and the Connectivity Map. The prognostic value of KRT14 was additionally corroborated through a comparison of protein levels in peritumoral and cancerous tissues via immunohistochemistry. Moreover, an immune-related prognostic model based on KRT14 was designed to enhance the prediction accuracy for the prognosis of BC patients. RESULTS The study found that KRT14 expression was generally downregulated in BC, correlating strongly with poor prognosis. Compared to normal tissues, the methylation level of KRT14 was higher in BC tissues. Lower expression of KRT14 was linked to decreased anti-tumoral immune cells infiltration and increased immunosuppressive cells infiltration. Sensitivity to various key therapeutic drugs was lower in groups with diminished KRT14 expression. In addition, several potential anti-BC small molecule compounds were identified. The model designed in this study significantly enhanced the predictive capability for BC patients compared to predictions based solely on KRT14 expression levels. CONCLUSION Overall, KRT14 was closely correlated with the prognosis in BC, making it a reliable biomarker.
Collapse
Affiliation(s)
- Siqi Liao
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xin Zhang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lanhui Chen
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jianning Zhang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Weiyu Lu
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Mengou Rao
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yifan Zhang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Zijian Ye
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Deyana Ivanova
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston MA02115, USA
| | - Fangfang Li
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Xuemei Chen
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Anchao Song
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Biao Xie
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| | - Meijiao Wang
- Department of Physiology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China; Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
7
|
Chang Y, Long M, Shan H, Liu L, Zhong S, Luo JL. Combining gut microbiota modulation and immunotherapy: A promising approach for treating microsatellite stable colorectal cancer. Crit Rev Oncol Hematol 2025; 208:104629. [PMID: 39864533 DOI: 10.1016/j.critrevonc.2025.104629] [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: 12/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and lethal cancers worldwide, ranking third in incidence and second in mortality. While immunotherapy has shown promise in patients with deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H), its effectiveness in proficient mismatch repair (pMMR) or microsatellite stable (MSS) CRC remains limited. Recent advances highlight the gut microbiota as a potential modulator of anti-tumor immunity. The gut microbiome can significantly influence the efficacy of immune checkpoint inhibitors (ICIs), especially in pMMR/MSS CRC, by modulating immune responses and systemic inflammation. This review explores the role of the gut microbiota in pMMR/MSS CRC, the mechanisms by which it may enhance immunotherapy, and current strategies for microbiota modulation. We discuss the potential benefits of combining microbiota-targeting interventions with immunotherapy to improve treatment outcomes for pMMR/MSS CRC patients.
Collapse
Affiliation(s)
- Yujie Chang
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Min Long
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Hanguo Shan
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Logen Liu
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Shangwei Zhong
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Jun-Li Luo
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, USC, Hunan 410008, China.
| |
Collapse
|
8
|
Chikaishi Y, Matsuoka H, Sugihara E, Takeda M, Sumitomo M, Yamada S, Inaguma G, Omura Y, Cheong Y, Kobayashi Y, Nakauchi M, Hiro J, Masumori K, Otsuka K, Nishihara H, Suda K, Saya H, Takimoto T. Mutation Analysis of TMB-High Colorectal Cancer: Insights Into Molecular Pathways and Clinical Implications. Cancer Sci 2025; 116:1082-1093. [PMID: 39822019 PMCID: PMC11967252 DOI: 10.1111/cas.16455] [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: 09/19/2024] [Revised: 01/06/2025] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
Colorectal cancer (CRC) is well characterized in terms of genetic mutations and the mechanisms by which they contribute to carcinogenesis. Mutations in APC, TP53, and KRAS are common in CRC, indicating key roles for these genes in tumor development and progression. However, for certain tumors with low frequencies of these mutations that are defined by tumor location and molecular phenotypes, a carcinogenic mechanism dependent on BRAF mutations has been proposed. We here analyzed targeted sequence data linked to clinical information for CRC, focusing on tumors with a high tumor mutation burden (TMB) in order to identify the characteristics of associated mutations, their relations to clinical features, and the mechanisms of carcinogenesis in tumors lacking the major driver oncogenes. Analysis of overall mutation frequencies confirmed that APC, TP53, and KRAS mutations were the most prevalent in our cohort. Compared with other tumors, TMB-high tumors were more frequent on the right side of the colon, had lower KRAS and higher BRAF mutation frequencies as well as a higher microsatellite instability (MSI) score, and showed a greater contribution of a mutational signature associated with MSI. Ranking of variant allele frequencies to identify genes that play a role early in carcinogenesis suggested that mutations in genes related to the DNA damage response (such as ATM and POLE) and to MSI (such as MSH2 and MSH6) may precede BRAF mutations associated with activation of the serrated pathway in TMB-high tumors. Our results thus indicate that TMB-high tumors suggest that mutations of genes related to mismatch repair and the DNA damage response may contribute to activation of the serrated pathway in CRC.
Collapse
Affiliation(s)
- Yuko Chikaishi
- Department of SurgeryFujita Health UniversityToyoakeAichiJapan
| | | | - Eiji Sugihara
- Research Promotion Headquarters, Open Facility CenterFujita Health UniversityToyoakeJapan
- Oncology Innovation CenterFujita Health UniversityToyoakeAichiJapan
| | - Mayu Takeda
- Faculty of Health and Medical SciencesAichi Syukutoku UniversityNagakuteAichiJapan
| | - Makoto Sumitomo
- Oncology Innovation CenterFujita Health UniversityToyoakeAichiJapan
| | - Seiji Yamada
- Oncology Innovation CenterFujita Health UniversityToyoakeAichiJapan
| | - Gaku Inaguma
- Department of SurgeryFujita Health UniversityToyoakeAichiJapan
| | - Yusuke Omura
- Department of SurgeryFujita Health UniversityToyoakeAichiJapan
| | | | | | - Masaya Nakauchi
- Department of Advanced Robotic and Endoscopic SurgeryFujita Health UniversityToyoakeAichiJapan
| | - Junichiro Hiro
- Department of SurgeryFujita Health UniversityToyoakeAichiJapan
| | - Koji Masumori
- Department of SurgeryFujita Health UniversityToyoakeAichiJapan
| | - Koki Otsuka
- Department of Advanced Robotic and Endoscopic SurgeryFujita Health UniversityToyoakeAichiJapan
| | - Hiroshi Nishihara
- Center for Cancer GenomicsKeio University School of MedicineTokyoJapan
| | - Koichi Suda
- Department of SurgeryFujita Health UniversityToyoakeAichiJapan
- Collaborative Laboratory for Research and Development in Advanced Surgical IntelligenceFujita Health UniversityToyoakeAichiJapan
| | - Hideyuki Saya
- Oncology Innovation CenterFujita Health UniversityToyoakeAichiJapan
| | - Tetsuya Takimoto
- Oncology Innovation CenterFujita Health UniversityToyoakeAichiJapan
| |
Collapse
|
9
|
Liu Z, Jiang X, Ke Z, Wang W, Tang J, Dai Y. PAR2 deficiency impairs antitumor immunity and attenuates anti-PD1 efficacy in colorectal cancer. Pharmacol Res 2025:107721. [PMID: 40174816 DOI: 10.1016/j.phrs.2025.107721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
A T cell-inflamed tumor microenvironment is predictive of better prognosis and clinical response to immunotherapy. Proteinase-activated receptor 2 (PAR2), a member of G-protein coupled receptors is involved in inflammatory process and the progression of various cancers. However, the role of PAR2 in modulating the tumor microenvironment remains unclear. Here, we found that PAR2 high-expression was associated with a favorable prognosis in patients with colorectal cancer. Intriguingly, PAR2 expression in human colorectal cancer was mainly confined to tumor cells and was significantly associated with CD8+ T cell infiltration. Tumor-intrinsic PAR2 deficiency blunted antitumor immune responses to promote tumor growth and attenuated the therapeutic efficacy of anti-PD1 in a mouse model of colon cancer. Tumors with downregulated PAR2 showed decreased CD8+ T cell infiltration and impaired effector function. Mechanistically, PAR2 activation in tumor cells induced CXCL9 and CXCL10 production via PI3K/AKT/mTOR signaling, thereby enhancing CD8+ T cell recruitment in the tumor microenvironment. In addition, PAR2 was essential for dendritic cell activation and differentiation towards conventional type 1 subset. PAR2 deficiency in dendritic cells markedly impaired their ability to prime CD8+ T cells and control tumor growth in vivo. Thus, our findings identify new roles for PAR2 in promoting antitumor immunity and provide a promising target to improve immunotherapy efficacy in colorectal cancer.
Collapse
Affiliation(s)
- Zilin Liu
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Xuehui Jiang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Ziliang Ke
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Weihong Wang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Jianqiang Tang
- Department of General Surgery, Peking University First Hospital, Beijing, China.
| | - Yun Dai
- Department of Gastroenterology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
10
|
He KJ, Gong G. Deciphering the integrated immunogenomic landscape of colorectal cancer: insights from Mendelian randomization and immune-stratified molecular subtyping. BMC Gastroenterol 2025; 25:213. [PMID: 40165100 PMCID: PMC11959972 DOI: 10.1186/s12876-025-03776-4] [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: 07/26/2024] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
PURPOSE This study aimed to decipher the intricate interplay between the immune landscape and CRC pathogenesis, elucidating how distinct immunophenotypes causally influence disease susceptibility and stratify patient outcomes. METHODS We obtained the immunocyte phenotypes and CRC data from their respective genome-wide association studies. The primary analysis used the inverse variance weighting (IVW) method. We also simultaneously employed MR-Egger, weighted mode, simple mode, and weighted median approaches to strengthen the findings. Consensus clustering stratified 619 TCGA CRC patients by immunome expression. Functional assays examined the tumor suppressor GPD1L. RESULTS The IVW MR analysis identified 17 immunocyte phenotypes positively potentially associated with increased CRC risk (P < 0.05, OR > 1), and 18 phenotypes negatively potentially associated with decreased CRC risk (P < 0.05, OR < 1). These associations were not confounded by heterogeneity or horizontal pleiotropy (P > 0.05). Reverse MR analysis further revealed 4 additional immunocyte phenotypes positively potentially associated with CRC (P < 0.05, OR > 1). Clustering resolved prognostic C1/C2 subtypes dependent on coordinated immunophenotypic programs. GPD1L knockdown promoted CRC cell proliferation. CONCLUSIONS Genetic interrogation delineated causal immunome-CRC relationships at single-cell resolution. Immune-stratified CRC subtyping stratified patient outcomes. GPD1L exhibited tumor-suppressive functions. Our findings establish an integrated immunogenomic framework elucidating CRC pathogenesis with implications for precision immunotherapies.
Collapse
Affiliation(s)
- Ke-Jie He
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou city, Zhejiang Province, China.
| | - Guoyu Gong
- School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
11
|
Wang SL, Chan TA. Navigating established and emerging biomarkers for immune checkpoint inhibitor therapy. Cancer Cell 2025:S1535-6108(25)00107-2. [PMID: 40154483 DOI: 10.1016/j.ccell.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
12
|
Liu H, Yang R, Zhong H, Zhang Y, Wang S, Guo K, Jiang Z, He J, Huang Y, Lin Y, Chen X, Lin J. Mechanism of Qingjie Fuzheng Granules in inhibiting colitis associated colorectal cancer by regulating TLR4 and IL-4R mediated macrophage polarization. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119511. [PMID: 39978444 DOI: 10.1016/j.jep.2025.119511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/14/2025] [Accepted: 02/15/2025] [Indexed: 02/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qingjie Fuzheng Granules (QFG), a herbal formula, has been employed as an adjuvant therapy for colitis-associated colorectal cancer (CAC), yet the underlying mechanisms by which QFG operates remain unclear. AIMS OF THE STUDY The aim of this study is to investigate whether the potential mechanism of QFG against CAC is associated with macrophage polarization. MATERIALS AND METHODS Non-targeted metabolomics and molecular docking assessed potential compounds of QFG to interact with targets associated with macrophage polarization. A model of AOM/DSS-induced CAC mice was established to analyze the effects of QFG on macrophage polarization using flow cytometry and immunohistochemical staining. In vitro experiments involved models of Ana-1 macrophages, either induced by varying QFG concentrations or with MD2 knockdown, to analyze M1-like phenotype. Meanwhile, M2-like macrophages models induced by IL-4 or culture supernatant of CT26 cells were utilized to assess the effects of QFG on M2-like macrophages. Finally, the mRNA expression of M1-like phenotype related to TLR4 pathways and the protein expression in IL-4R-mediated pathways were analyzed using RT-qPCR and Western blot, respectively. RESULTS Molecular docking confirmed the presence of binding sites between the ingredients of QFG and IL-4R or TLR4/MD2 receptor complex. QFG could induce a shift in macrophages towards an M1-like phenotype while inhibiting an M2-like phenotype in the colon with CAC mice and Ana-1 macrophages. QFG resulted in the upregulation of iNOS, IL-6, IL-1β, and TNF-α mRNA expression, which could be counteracted by TAK242, SR11302, INH14, PDTC, and LY294002, or by the knockdown of MD2. Meanwhile, QFG inhibited IL-4R-induced phosphorylation of STAT 6 and Akt. CONCLUSION Various monomer components within QFG can bind to MD2 or IL-4R, respectively, thereby inducing macrophages towards an M1-like phenotype through TLR4-mediated NF-κB, MAPK, and PI3K/Akt pathway activation, or inhibiting macrophages towards an M2-like phenotype via IL-4R-mediated JAKs pathway inhibition, ultimately exerting an inhibitory effect on the occurrence and development of CAC.
Collapse
Affiliation(s)
- Haiqin Liu
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China; Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, Fujian Province, 350122, China
| | - Ruiming Yang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China; Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, Fujian Province, 350122, China
| | - Hangyan Zhong
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China; Department of Proctology, Shanghang Hospital of Traditional Chinese Medicine, Longyan, Fujian Province, 364200, China
| | - Youquan Zhang
- The Second People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350001, China
| | - Shunyong Wang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China
| | - Kangyue Guo
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China
| | - Zhishan Jiang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China
| | - Jiajun He
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China
| | - Yunmei Huang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China; Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, Fujian Province, 350122, China
| | - Ying Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, 350001, China; Department of Pathology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian Province, 350001, China
| | - Xuzheng Chen
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China; Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, Fujian Province, 350122, China.
| | - Jiumao Lin
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China; Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, Fujian Province, 350122, China.
| |
Collapse
|
13
|
Zhang J, Liu G, Wang W. PRSS53 is a potential novel biomarker related to prognosis and immunity in clear cell renal cell carcinoma. Discov Oncol 2025; 16:362. [PMID: 40111561 PMCID: PMC11925835 DOI: 10.1007/s12672-025-02114-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE To analyze the expression levels, clinical significance, Immune infiltration and prognostic value of PRSS53 (Protease Serine 53) in clear cell renal cell carcinoma (ccRCC) using bioinformatics methods. METHODS Data on PRSS53 in ccRCC were extracted from databases and platforms, including The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression Project (GTEx), The Gene Expression Omnibus (GEO), Xiantao Academic Tool, Human Protein Atlas (HPA) and so on. We analyzed the relationship between PRSS53 expression and the clinical and pathological characteristics, diagnosis, immune infiltration and prognosis in ccRCC patients. Additionally, immunohistochemical analysis was performed on 9 pairs of ccRCC patient samples. RESULTS PRSS53 was significantly upregulated in ccRCC and was closely associated with the TNM stage and histological grade of ccRCC. Receiver operating characteristic (ROC) curve analysis demonstrated the excellent diagnostic performance of PRSS53 in ccRCC (AUC = 0.928). Patients with high PRSS53 expression exhibited lower overall survival (OS) and disease-specific survival (DSS). Gene set enrichment analysis (GSEA) revealed that PRSS53 is involved in cellular functions such as anchored component of membrane, basement membrane and RNA-binding involved in post-transcriptional gene silencing. Single-sample GSEA (ssGSEA) indicated a positive correlation between PRSS53 expression and T helper cells infiltration levels, and a negative correlation with T gamma delta (Tgd) cell infiltration. PRSS53 was predominantly expressed in renal proximal tubules. The immunohistochemical results and HPA database showed that PRSS53 protein expression was significantly lower in clinical ccRCC tissues compared to normal tissues. CONCLUSION PRSS53 is a new prognostic biomarker and potential therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Jiajun Zhang
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224006, China
| | - Guocheng Liu
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224006, China
| | - Wei Wang
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224006, China.
- Department of Urology, Yancheng No.1 People's Hospital, Yancheng, 224006, China.
| |
Collapse
|
14
|
Zhang H, Chen L, Li L, Liu Y, Das B, Zhai S, Tan J, Jiang Y, Turco S, Yao Y, Frishman D. Prediction and analysis of tumor infiltrating lymphocytes across 28 cancers by TILScout using deep learning. NPJ Precis Oncol 2025; 9:76. [PMID: 40108446 PMCID: PMC11923303 DOI: 10.1038/s41698-025-00866-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
The density of tumor-infiltrating lymphocytes (TILs) serves as a valuable indicator for predicting anti-tumor responses, but its broad impact across various types of cancers remains underexplored. We introduce TILScout, a pan-cancer deep-learning approach to compute patch-level TIL scores from whole slide images (WSIs). TILScout achieved accuracies of 0.9787 and 0.9628, and AUCs of 0.9988 and 0.9934 in classifying WSI patches into three categories-TIL-positive, TIL-negative, and other/necrotic-on validation and independent test sets, respectively, surpassing previous studies. The biological significance of TILScout-derived TIL scores across 28 cancers was validated through comprehensive functional and correlational analyses. A consistent decrease in TIL scores with an increase in cancer stage provides direct evidence that the lower TIL content may stimulate cancer progression. Additionally, TIL scores correlated with immune checkpoint gene expression and genomic variation in common cancer driver genes. Our comprehensive pan-cancer survey highlights the critical prognostic significance of TILs within the tumor microenvironment.
Collapse
Affiliation(s)
- Huibo Zhang
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lulu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Liu
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Barnali Das
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Shuang Zhai
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Juan Tan
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan Jiang
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Simona Turco
- Electrical Engineering, Eindhoven University of Technology, Den Dolech 12, Eindhoven, 5612AZ, the Netherlands
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Dmitrij Frishman
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
15
|
Long C, Cheng J, Feng M, Yan B, Li Y, Jiang W, Chen D, Yan J. Association of the tumor microenvironment collagen score and immunoscore with colon cancer lymph node metastasis. BMC Cancer 2025; 25:506. [PMID: 40108544 PMCID: PMC11924846 DOI: 10.1186/s12885-025-13842-5] [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: 05/05/2024] [Accepted: 02/28/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND In clinical practice, lymph node status has an important impact on colon cancer (CC) management and treatment. The role of the tumor microenvironment collagen score and immunoscore in colon cancer lymph node metastasis remains unknown. METHODS A total of 249 CC patients who underwent laparoscopic-assisted D3 lymphadenectomy from June 2016 to May 2019 were included. The patients' clinicopathological data were collected retrospectively. A total of 142 collagen features were extracted by multiphoton imaging and collagen quantification. A collagen score was constructed using a LASSO logistic regression model. Antibodies against CD3 and CD8 were used for immunostaining. The immunoscore was constructed based on the mean densities of CD3 + and CD8 + T cells both in the tumor center and invasion margin on imaging. RESULTS The lymph node metastasis rate among colon cancer patients was 42.2% (105/249). The multivariate analysis indicated that lymphatic invasion (OR: 3.892, 95% CI: 1.784-8.491, p = 0.001), vascular invasion (OR, 3.234, 95% CI: 1.544-6.776); p = 0.002), mucus adenocarcinoma and signet-ring cell carcinoma (OR: 2.990, 95% CI: 1.413-6.328, p = 0.004), the collagen score (OR: 6.304, 95% CI: 2.145-18.527, p = 0.001) and the immunoscore [intermediate group (OR, 2.473; 95% CI, 1.192-5.130; p = 0.015); low group (OR, 5.877; 95% CI, 2.423-14.257; p < 0.01)] were independent risk factors for colon cancer lymph node metastasis. The newly developed model comprising these five independent predictors showed good discrimination with an AUROC of 0.809 (95% CI: 0.755-0.862). The new model performed significantly better than the traditional clinicopathological model [AUROC: 0.715 (95% CI: 0.649-0.780), p < 0.001]. CONCLUSIONS The tumor microenvironment collagen score and immunoscore are associated with colon cancer lymph node metastasis.
Collapse
Affiliation(s)
- Chenyan Long
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Guangzhou, Guangdong, 510515, People's Republic of China
- Department of Colorectal & Anal Surgery, Cancer Hospitalaffiliated to, Guangxi Medical Universityaq , Nanning, Guangxi, 530021, People's Republic of China
| | - Jiaxin Cheng
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Mingyuan Feng
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Botao Yan
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Yiran Li
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Wei Jiang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Dexin Chen
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Jun Yan
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China.
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China.
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Guangzhou, Guangdong, 510515, People's Republic of China.
| |
Collapse
|
16
|
Takahashi H, Hanaoka K, Wada H, Kojima D, Watanabe M. The Current Status of T Cell Receptor (TCR) Repertoire Analysis in Colorectal Cancer. Int J Mol Sci 2025; 26:2698. [PMID: 40141338 PMCID: PMC11943327 DOI: 10.3390/ijms26062698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Abstract
The rapid increase in colorectal cancer (CRC) cases recently has highlighted the need to use predictive biomarkers to guide therapeutic approaches. Current studies have focused on the tumor-infiltrating lymphocytes present in the tumor microenvironment (TME), in which cytotoxic T cell activation and the amount are associated with CRC patient prognosis. The T cell receptor (TCR) is essential for antigen recognition and T cell identification, playing a central role in cancer immunotherapy. The T cell status reflects TCR diversity or clonality, known as the TCR repertoire. Accordingly, analyzing the TCR repertoire dynamics may help predict the immunological circumstances of the TME in a timely way. In this review, we summarize the TCR repertoire-related knowledge, including its potential use as predictive biomarkers in CRC. The intratumoral TCR repertoire is restricted in CRC patients compared with healthy individuals, as well as in peripheral blood. Patients with deficient mismatch repair display more restriction than those with proficient mismatch repair. Importantly, a higher TCR diversity before treatment and a decrease following treatment may indicate a good response and a better clinical outcome in CRC patients. The future use of TCR repertoire sequencing technology combined with artificial intelligence-based analysis is a potential strategy for CRC therapeutic decision making.
Collapse
Affiliation(s)
- Hiroyuki Takahashi
- Department of Surgery, Fukuoka University Chikushi Hospital, 1-1-1 Zokumyoin, Chikushino 818-8502, Fukuoka, Japan; (K.H.); (H.W.); (D.K.); (M.W.)
| | | | | | | | | |
Collapse
|
17
|
Zhang X, Liu S, Wu K, Shu L, Li Y, Li L, Wang D. Structural characteization and anti-colorectal cancer activity of a fucogalactan purified from Ganoderma tsugae. Carbohydr Polym 2025; 352:123203. [PMID: 39843104 DOI: 10.1016/j.carbpol.2024.123203] [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: 11/07/2024] [Revised: 12/25/2024] [Accepted: 12/29/2024] [Indexed: 01/24/2025]
Abstract
Ganoderma tsugae, a traditional medicinal mushroom, exhibits anti-tumor properties; however, the effects of its polysaccharide on anti-colorectal cancer remain undetermined. Herein, a fucogalactan of Ganoderma tsugae (GTP-a2) was isolated and purified from its fruiting body. The molecular weight of GTP-a2 is 7.056 kDa, consisting of →6)-α-D-Galp-(1→ backbone with branches of α-L-Fucp-(1→, which is attached at C2. Subsequently, the anti-colorectal cancer activity and potential mechanism of GTP-a2 were investigated in azoxymethane/dextran sulfate sodium (AOM/DSS)-induced colitis-associated colorectal cancer (CAC) mice. GTP-a2 reduced colorectal tumor numbers and suppressed tumor development. Metabolite analysis of the colon revealed that GTP-a2 altered cancer-related metabolites, notably increasing ophiobolin A level. Combined with proteomics and biochemical detection data revealed that GTP-a2 regulated the levels of Aldh1a3 through the mammalian target of rapamycin (mTOR)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway in vivo and in vitro. Additionally, GTP-a2 regulated immune function by inhibiting macrophage polarization to M1-like phenotype. These results suggest the potential application of GTP-a2 as a therapeutic agent for CAC.
Collapse
Affiliation(s)
- Xin Zhang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China.
| | - Shuai Liu
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection and Mycology, Jilin Agricultural University, Changchun 130118, China.
| | - Keyi Wu
- College of Plant Protection and Mycology, Jilin Agricultural University, Changchun 130118, China.
| | - Lili Shu
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China.
| | - Yu Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China; College of Plant Protection and Mycology, Jilin Agricultural University, Changchun 130118, China.
| | - Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection and Mycology, Jilin Agricultural University, Changchun 130118, China.
| | - Di Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; College of Plant Protection and Mycology, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
18
|
Trailin A, Ali E, Ye W, Pavlov S, Červenková L, Vyčítal O, Ambrozkiewicz F, Hošek P, Daum O, Liška V, Hemminki K. Prognostic assessment of T-cells in primary colorectal cancer and paired synchronous or metachronous liver metastasis. Int J Cancer 2025; 156:1282-1292. [PMID: 39508720 PMCID: PMC11736993 DOI: 10.1002/ijc.35252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024]
Abstract
Prognostic value of T-cells between primary colorectal cancer (pCRC) and its paired synchronous and metachronous liver metastasis (LM) is underinvestigated and is the subject of the present study. We enrolled into this retrospective cohort study patients, who underwent resection of both pCRC and synchronous LM (N = 55) or metachronous LM (N = 44). After immunohistochemical staining for CD3+, CD8+, and CD45R0+ whole slides were scanned and T-cell densities were quantified using QuPath software in tumor center (TC), inner margin (IM), outer margin (OM), and peritumor zone (PT) of pCRC and LM. High densities of CD8+ T-cells in TC, OM and PT of synchronous LM were associated with longer disease-free survival (DFS). Greater densities of CD3+ T-cells in IM and PT and CD8+ T-cells in IM, OM and PT in synchronous LM over pCRC were associated with longer DFS. Greater densities of CD8+ T-cells in the TC and IM and CD3+ T-cells in the IM of pCRC were found in the metachronous over synchronous group. The first novel finding demonstrated that high density of CD8+ T cells in synchronous LM were associated with favorable outcome. The second finding of high CD8+ cell density in pCRC in metachronous over synchronous CRC may provide a mechanistic basis for the delay of metastatic spread. Both findings could be applied clinically with own reference values.
Collapse
Affiliation(s)
- Andriy Trailin
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Esraa Ali
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Wenjing Ye
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Sergii Pavlov
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Lenka Červenková
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Ondřej Vyčítal
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Filip Ambrozkiewicz
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Petr Hošek
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Ondřej Daum
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Václav Liška
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | - Kari Hemminki
- Laboratory of Translational Cancer Genomics, Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| |
Collapse
|
19
|
Wang Y, Anesi JC, Panicker IS, Cook D, Bista P, Fang Y, Oqueli E. Neuroimmune Interactions and Their Role in Immune Cell Trafficking in Cardiovascular Diseases and Cancer. Int J Mol Sci 2025; 26:2553. [PMID: 40141195 PMCID: PMC11941982 DOI: 10.3390/ijms26062553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Sympathetic nerves innervate bone marrow and various immune organs, where norepinephrine-the primary sympathetic neurotransmitter-directly interacts with immune cells that express adrenergic receptors. This article reviewed the key molecular pathways triggered by sympathetic activation and explored how sympathetic activity influences immune cell migration. Norepinephrine serves as a chemoattractant for monocytes, macrophages, and stem cells, promoting the migration of myeloid cells while inhibiting the migration of lymphocytes at physiological concentrations. We also examined the role of immune cell infiltration in cardiovascular diseases and cancer. Evidence suggests that sympathetic activation increases myeloid cell infiltration into target tissues across various cardiovascular diseases, including atherosclerosis, hypertension, cardiac fibrosis, cardiac hypertrophy, arrhythmia, myocardial infarction, heart failure, and stroke. Conversely, inhibiting sympathetic activity may serve as a potential therapeutic strategy to treat these conditions by reducing macrophage infiltration. Furthermore, sympathetic activation promotes macrophage accumulation in cancer tissues, mirroring its effects in cardiovascular diseases, while suppressing T lymphocyte infiltration into cancerous sites. These changes contribute to increased cancer growth and metastasis. Thus, inhibiting sympathetic activation could help to protect against cancer by enhancing T cell infiltration and reducing macrophage presence in tumors.
Collapse
Affiliation(s)
- Yutang Wang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Jack C. Anesi
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Indu S. Panicker
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Darcy Cook
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Prapti Bista
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Yan Fang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Ernesto Oqueli
- Cardiology Department, Grampians Health Ballarat, Ballarat, VIC 3353, Australia
- School of Medicine, Faculty of Health, Deakin University, Geelong, VIC 3217, Australia
| |
Collapse
|
20
|
Silinskaite U, Valciukiene J, Jakubauskas M, Poskus T. The Immune Environment in Colorectal Adenoma: A Systematic Review. Biomedicines 2025; 13:699. [PMID: 40149674 PMCID: PMC11940254 DOI: 10.3390/biomedicines13030699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Research on colorectal adenoma is significantly less comprehensive compared to studies on colorectal carcinoma. Although colorectal adenoma is a precursor of the majority of sporadic colorectal cancers, not all adenomas develop into carcinomas. The complex interaction of immune responses in the premalignant tumor microenvironment might be a factor for that. Methods: In this systematic review, we aim to provide a thorough analysis of the current research examining the immune infiltration patterns in sporadic colorectal adenoma tissues in the context of immune cell-based, cytokine-based, and other immunological factor-related changes along the conventional adenoma-carcinoma sequence. The articles included in the review extend up to December 2024 in PubMed and Web of Science databases. Results: Most included studies have shown significant differences in immune cell counts, densities, and cytokine expression levels associated with premalignant colorectal lesions (and/or colorectal cancer). No consensus on the immune-related tendencies concerning CD4+T cells and CD8+T cells was reached. Decreasing expression of mDCs and plasma and naïve B cells were detected along the ACS. The increased density of tissue eosinophils in the adenoma tissue dramatically diminishes after the transition to carcinoma. As the adenoma progresses, the increasing expression of IL-1α, IL-4, IL-6, IL-8, IL-10, IL-17A, IL-21, IL-23, IL-33, and TGF-β and decreasing levels of IL-12A, IL-18, IFN-γ, and TNFα cytokines in the invasive carcinoma stage is being detected. The over-expression of COX-2, PD-1/PD-L1, CTLA-4, and ICOS/ICOSLG in the colorectal adenomatous and cancerous tissues was also observed. Conclusions: Further studies are needed for a better understanding of the whole picture of colorectal adenoma-associated immunity and its impact on precancerous lesion's potential to progress.
Collapse
|
21
|
Imaoka K, Shimomura M, Okuda H, Yano T, Shimizu W, Yoshimitsu M, Ikeda S, Nakahara M, Kohyama M, Kobayashi H, Shimizu Y, Kochi M, Sumitani D, Mukai S, Takakura Y, Ishizaki Y, Kodama S, Fujimori M, Ishikawa S, Adachi T, Ohdan H. Multivisceral resection as a key indicator of recurrence in locally advanced colorectal cancers with pathologic T3 tumors. J Gastrointest Surg 2025; 29:102015. [PMID: 40081790 DOI: 10.1016/j.gassur.2025.102015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/23/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
PURPOSE This study aimed to elucidate the clinical outcomes of patients with pathologic T3 (pT3) and pathologic T4 (pT4) tumors who underwent radical resection with multivisceral resection (MVR) and to assess the prognostic significance of MVR in locally advanced colorectal cancers (CRCs) in pT3 and pT4 tumors. METHODS This multicenter retrospective analysis evaluated the characteristics, clinicopathologic stages, perioperative factors, and clinical outcomes of patients who underwent primary colorectal resection. Patients were divided into 4 groups: those with a pT3 tumor who did not undergo MVR (pT3 - MVR; n = 1108), those with a pT3 tumor who underwent MVR (pT3 + MVR; n = 56), those with a pT4 tumor who did not undergo MVR (pT4 - MVR; n = 306), and those with a pT4 tumor who did underwent MVR (pT4 + MVR; n = 123). Univariate and multivariate regression analyses were performed to identify risk factors for recurrence. RESULTS The pT3 + MVR group exhibited a higher 5-year recurrence rate than the pT3 - MVR group, with recurrence rates similar to those of the pT4 - MVR or pT4 + MVR groups (pT3 - MVR, 17.4%; pT3 + MVR, 31.6%; pT4 - MVR, 33.4%; pT4 + MVR, 35.1%). Multivariate analysis identified MVR as an independent risk factor for recurrence, particularly peritoneal dissemination, in pT3 tumors, whereas MVR had less effect on recurrence in pT4 tumors. CONCLUSION pT3 tumors requiring MVR had a higher recurrence rate than pT4 tumors. The surgeon's clinical assessment of potential T4 tumors requiring MVR at the time of surgery was an important prognostic indicator in advanced CRC.
Collapse
Affiliation(s)
- Kouki Imaoka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Manabu Shimomura
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Hiroshi Okuda
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takuya Yano
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Wataru Shimizu
- Department of Surgery, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Masanori Yoshimitsu
- Department of Surgery, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Satoshi Ikeda
- Department of Gastroenterological Surgery, Hiroshima Prefectural Hospital, Hiroshima, Japan
| | | | - Mohei Kohyama
- Department of Surgery, Hiroshima General Hospital, Hatsukaichi, Japan
| | | | - Yosuke Shimizu
- Department of Surgery, Kure Medical Center/Chugoku Cancer Center, Institute for Clinical Research, Kure, Japan
| | - Masatoshi Kochi
- Department of Surgery, National Hospital Organization Higashihiroshima Medical Center, Higashihiroshima, Japan
| | | | | | - Yuji Takakura
- Department of Surgery, Chuden Hospital, Hiroshima, Japan
| | - Yasuyo Ishizaki
- Department of Surgery, National Hospital Organization Hiroshima-Nishi Medical Center, Otake, Japan
| | - Shinya Kodama
- Department of Surgery, Yoshida General Hospital, Akitakata, Japan
| | - Masahiko Fujimori
- Department of Surgery, Kure Medical Association Hospital, Kure, Japan
| | - Sho Ishikawa
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomohiro Adachi
- Department of Surgery, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
22
|
Ueshima E, Sofue K, Komatsu S, Ishihara N, Komatsu M, Umeno A, Nishiuchi K, Kozuki R, Yamaguchi T, Matsuura T, Tada T, Murakami T. Immunoscore Predicted by Dynamic Contrast-Enhanced Computed Tomography Can Be a Non-Invasive Biomarker for Immunotherapy Susceptibility of Hepatocellular Carcinoma. Cancers (Basel) 2025; 17:948. [PMID: 40149284 PMCID: PMC11940361 DOI: 10.3390/cancers17060948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 02/21/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Although immunotherapy is the primary treatment option for intermediate-stage hepatocellular carcinoma (HCC), its efficacy varies. This study aimed to identify non-invasive imaging biomarkers predictive of the immunoscore linked to dynamic contrast-enhanced computed tomography (CECT). Methods: We performed immunohistochemical staining with CD3+ and CD8+ antibodies and counted the positive cells in the invasive margin (IM) and central tumor (CT), converting them to an immunoscore of 0 to 4 points. We assessed the dynamic CECT findings obtained from 96 patients who underwent hepatectomy for HCC and evaluated the relationship between dynamic CECT findings and immunoscores. For validation, we assessed the treatment effects on 81 nodules using the Response Evaluation Criteria in Solid Tumors in another cohort of 41 patients who received combined immunotherapy with atezolizumab and bevacizumab (n = 27) and durvalumab and tremelizumab (n = 14). Results: HCCs with peritumoral enhancement in the arterial phase (p < 0.001) and rim APHE (p = 0.009) were associated with the immunoscore in univariate linear regression analysis and peritumoral enhancement in the arterial phase (p = 0.004) in multivariate linear regression analysis. The time to nodular progression in HCCs with peritumoral enhancement in the arterial phase was significantly longer than that in HCCs without this feature (p < 0.001). Conclusions: We identified HCCs with peritumoral enhancement in the arterial phase as a noninvasive imaging biomarker to predict immune-inflamed HCC with a high immunoscore tendency. These HCCs were most likely to respond to combined immunotherapy.
Collapse
Affiliation(s)
- Eisuke Ueshima
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan; (E.U.); (K.N.); (R.K.); (T.M.)
| | - Keitaro Sofue
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan; (E.U.); (K.N.); (R.K.); (T.M.)
| | - Shohei Komatsu
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan; (S.K.); (N.I.)
| | - Nobuaki Ishihara
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan; (S.K.); (N.I.)
| | - Masato Komatsu
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan;
| | - Akihiro Umeno
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan; (E.U.); (K.N.); (R.K.); (T.M.)
| | - Kentaro Nishiuchi
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan; (E.U.); (K.N.); (R.K.); (T.M.)
| | - Ryohei Kozuki
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan; (E.U.); (K.N.); (R.K.); (T.M.)
| | - Takeru Yamaguchi
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan; (E.U.); (K.N.); (R.K.); (T.M.)
| | - Takanori Matsuura
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan; (T.M.); (T.T.)
| | - Toshifumi Tada
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan; (T.M.); (T.T.)
| | - Takamichi Murakami
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe 650-0017, Hyogo, Japan; (E.U.); (K.N.); (R.K.); (T.M.)
| |
Collapse
|
23
|
Han SH, Ju MH, Pak MG. Prognostic and therapeutic potential of CXCR6 expression on CD8 + T cells in gastric cancer: a retrospective cohort study. BMC Gastroenterol 2025; 25:139. [PMID: 40050760 PMCID: PMC11884069 DOI: 10.1186/s12876-025-03735-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/26/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a pressing global health concern, with prognosis intricately linked to the tumour stage and tumour microenvironment, especially, the presence of immune cells. Notably, CD8 + T cells play a pivotal role in the anti-tumour immune response, prompting investigations into their correlation with GC survival. This study aimed to investigate the intricate interplay between CD8 + T cells, particularly within the context of CXCR6, and survival outcomes in patients with GC. METHODS Utilising datasets from The Cancer Genome Atlas, Gene Expression Omnibus, and Tumor Immune Dysfunction and Exclusion, the study employed xCell and Weighted Gene Co-expression Network Analysis to assess CD8 + T cell infiltration and identify key gene clusters. The prognostic significance of CXCR6 was evaluated via immunohistochemical staining of a GC tissue microarray. RESULTS High CD8 + T cell infiltration correlated with improved survival in patients with GC. CXCR6 was identified as a prognostic gene and its expression was predominantly observed in CD8 + T cells. CXCR6 expression positively correlated with improved overall and disease-free survival. Furthermore, CXCR6 expression was associated with an immunoreactive microenvironment. CONCLUSION This study established that high CD8 + T-cell infiltration is related to CXCR6 expression, making it a key factor in predicting a favorable GC prognosis. The role of CXCR6 in shaping the tumour microenvironment and its potential utility in immunotherapy response prediction highlights its significance in GC management.
Collapse
Affiliation(s)
- Song-Hee Han
- Department of Pathology, Dong-A University College of Medicine, Busan, 49201, Republic of Korea.
| | - Mi Ha Ju
- Department of Pathology, Dong-A University College of Medicine, Busan, 49201, Republic of Korea
| | - Min Gyoung Pak
- Department of Pathology, Dong-A University College of Medicine, Busan, 49201, Republic of Korea
| |
Collapse
|
24
|
Lin H, Hua J, Wang Y, Chen M, Liang Y, Yan L, Zhao W, Luo S, Hong D, Chen X, Pan X, Liu J, Liu Z. Prognostic and predictive values of a multimodal nomogram incorporating tumor and peritumor morphology with immune status in resectable lung adenocarcinoma. J Immunother Cancer 2025; 13:e010723. [PMID: 40050046 PMCID: PMC11887283 DOI: 10.1136/jitc-2024-010723] [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: 10/02/2024] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Current prognostic and predictive biomarkers for lung adenocarcinoma (LUAD) predominantly rely on unimodal approaches, limiting their characterization ability. There is an urgent need for a comprehensive and accurate biomarker to guide individualized adjuvant therapy decisions. METHODS In this retrospective study, data from patients with resectable LUAD (stage I-III) were collected from two hospitals and a publicly available dataset, forming a training dataset (n=223), a validation dataset (n=95), a testing dataset (n=449), and the non-small cell lung cancer (NSCLC) Radiogenomics dataset (n=59). Tumor and peritumor scores were constructed from preoperative CT radiomics features (shape/intensity/texture). An immune score was derived from the density of tumor-infiltrating lymphocytes (TILs) within the cancer epithelium and stroma on hematoxylin and eosin-stained whole-slide images. A clinical score was constructed based on clinicopathological risk factors. A Cox regression model was employed to integrate these scores, thereby constructing a multimodal nomogram to predict disease-free survival (DFS). The adjuvant chemotherapy benefit rate was subsequently calculated based on this nomogram. RESULTS The multimodal nomogram outperformed each of the unimodal scores in predicting DFS, with a C-index of 0.769 (vs 0.634-0.731) in the training dataset, 0.730 (vs 0.548-0.713) in the validation dataset, and 0.751 (vs 0.660-0.692) in the testing dataset. It was independently associated with DFS after adjusting for other clinicopathological risk factors (training dataset: HR=3.02, p<0.001; validation dataset: HR=2.33, p<0.001; testing dataset: HR=2.03, p=0.001). The adjuvant chemotherapy benefit rate effectively distinguished between patients benefiting from adjuvant chemotherapy and those from observation alone (interaction p<0.001). Furthermore, the high-/low-risk groups defined by the multimodal nomogram provided refined stratification of candidates for adjuvant chemotherapy identified by current guidelines (p<0.001). Gene set enrichment analyses using the NSCLC Radiogenomics dataset revealed associations between tumor/peritumor scores and pathways involved in epithelial-mesenchymal transition, angiogenesis, IL6-JAK-STAT3 signaling, and reactive oxidative species. CONCLUSION The multimodal nomogram, which incorporates tumor and peritumor morphology with anti-tumor immune response, provides superior prognostic accuracy compared with unimodal scores. Its defined adjuvant chemotherapy benefit rates can inform individualized adjuvant therapy decisions.
Collapse
Affiliation(s)
- Huan Lin
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Junjie Hua
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yumeng Wang
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, Guangxi, China
| | - Mingwei Chen
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, Guangxi, China
| | - Yanting Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - LiXu Yan
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Zhao
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shiwei Luo
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Deqing Hong
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, Guangdong, China
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xipeng Pan
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, Guangxi, China
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, Guangdong, China
| |
Collapse
|
25
|
Chen Q, Zhao B, Tan Z, Hedberg G, Wang J, Gonzalez L, Mugabo CH, Johnsson A, Negrini E, Páez LP, Rodriguez L, James A, Chen Y, Mikeš J, Bernhardsson AK, Reitzner SM, von Walden F, O'Neill O, Barcenilla H, Wang C, Davis MM, Carlson LM, Pal N, Blomgren K, Repsilber D, Herold N, Lakshmikanth T, Kogner P, Ljungblad L, Brodin P. Systems-level immunomonitoring in children with solid tumors to enable precision medicine. Cell 2025; 188:1425-1440.e11. [PMID: 39837329 DOI: 10.1016/j.cell.2024.12.014] [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: 10/26/2023] [Revised: 10/20/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025]
Abstract
Cancer is the leading cause of death from disease in children. Survival depends not only on surgery, cytostatic drugs, and radiation but also on systemic immune responses. Factors influencing these immune responses in children of different ages and tumor types are unknown. Novel immunotherapies can enhance anti-tumor immune responses, but few children have benefited, and markers of effective responses are lacking. Here, we present a systems-level analysis of immune responses in 191 children within a population-based cohort with diverse tumors and reveal that age and tumor type shape immune responses differently. Systemic inflammation and cytotoxic T cell responses correlate with tumor mutation rates and immune cell infiltration. Clonally expanded T cell responses are rarely detected in blood or tumors at diagnosis but are sometimes elicited during treatment. Expanded T cells are similarly regulated in children and adults with more immunogenic cancers. This research aims to facilitate the development of precision immunotherapies for children with cancer.
Collapse
Affiliation(s)
- Qi Chen
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Binbin Zhao
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Ziyang Tan
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Gustav Hedberg
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Jun Wang
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Laura Gonzalez
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Constantin Habimana Mugabo
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Anette Johnsson
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Erika Negrini
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Laura Piñero Páez
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Lucie Rodriguez
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Anna James
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Yang Chen
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Jaromír Mikeš
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Anna Karin Bernhardsson
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Stefan Markus Reitzner
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden; Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ferdinand von Walden
- Neuropediatric Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Olivia O'Neill
- Department of Immunology and Inflammation, Imperial College London, London W12 EH7, UK; Medical Research Council, Laboratory of Medical Sciences, Imperial College Hammersmith Campus, London, UK
| | - Hugo Barcenilla
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Chunlin Wang
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark M Davis
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lena-Maria Carlson
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden; Section Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Niklas Pal
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden; Section Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Klas Blomgren
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden; Section Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Dirk Repsilber
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Nikolas Herold
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden; Section Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Tadepally Lakshmikanth
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden; Department of Immunology and Inflammation, Imperial College London, London W12 EH7, UK; Medical Research Council, Laboratory of Medical Sciences, Imperial College Hammersmith Campus, London, UK
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden; Section Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Linda Ljungblad
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Petter Brodin
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden; Department of Immunology and Inflammation, Imperial College London, London W12 EH7, UK; Medical Research Council, Laboratory of Medical Sciences, Imperial College Hammersmith Campus, London, UK; Pediatric Rheumatology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden.
| |
Collapse
|
26
|
Lehmann J, Thelen M, Kreer C, Schran S, Garcia-Marquez MA, Cisic I, Siepmann K, Hagen EM, Eckel HNC, Lohneis P, Kruger S, Boeck S, Ormanns S, Rudelius M, Werner J, Popp F, Klein F, von Bergwelt-Baildon MS, Bruns CJ, Quaas A, Wennhold K, Schlößer HA. Tertiary Lymphoid Structures in Pancreatic Cancer are Structurally Homologous, Share Gene Expression Patterns and B-cell Clones with Secondary Lymphoid Organs, but Show Increased T-cell Activation. Cancer Immunol Res 2025; 13:323-336. [PMID: 39661055 DOI: 10.1158/2326-6066.cir-24-0299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/02/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Tertiary lymphoid structures (TLS) in cancer are considered ectopic hotspots for immune activation that are similar to lymphoid follicles in secondary lymphoid organs (SLO). This study elucidates shared and TLS/SLO-specific features in pancreatic ductal adenocarcinoma (PDAC). TLS abundance was related to superior survival and T-cell abundance in 110 treatment-naïve PDAC samples, underlining their clinical relevance. Immunofluorescence microscopy identified structural homologies between TLSs and SLOs. In RNA expression analyses of laser-microdissected TLSs and paired SLOs, we observed largely overlapping expression patterns of immune-related gene clusters but distinct expression patterns of T-cell and complement-associated genes. Immune cells in TLS expressed essential markers of germinal center formation. Increased activation of tumor-draining lymph nodes in patients with high numbers of TLSs highlights the relevance of these tumor-related structures to systemic immune response. In line with this, we identified an overlap of expanded B-cell receptor clonotypes in TLSs and SLOs, which suggests a vivid cross-talk between the two compartments. We conclude that combined therapeutic approaches exploiting TLS-mediated antitumor immune responses may improve susceptibility of PDAC to immunotherapy.
Collapse
Affiliation(s)
- Jonas Lehmann
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Martin Thelen
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Christoph Kreer
- Laboratory of Experimental Immunology, Faculty of Medicine and University Hospital Cologne, Institute of Virology, University of Cologne Cologne, Germany
| | - Simon Schran
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Maria A Garcia-Marquez
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Igor Cisic
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Klara Siepmann
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Elena M Hagen
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Hans Nikolaus Caspar Eckel
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Cologne, Cologne, Germany
| | - Philipp Lohneis
- Institute of Pathology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Stephan Kruger
- Department of Internal Medicine III, University Hospital, Ludwig Maximilians University, Munich, Germany
| | - Stefan Boeck
- Department of Internal Medicine III, University Hospital, Ludwig Maximilians University, Munich, Germany
- Department of Hematology and Oncology, München Klinik Neuperlach, Munich, Germany
| | - Steffen Ormanns
- Faculty of Medicine, Institute of Pathology, Ludwig Maximilians University, Munich, Germany
- Innpath Institute of Pathology, Tirol Kliniken, Innsbruck, Austria
| | - Martina Rudelius
- Faculty of Medicine, Institute of Pathology, Ludwig Maximilians University, Munich, Germany
| | - Jens Werner
- Department of General, Visceral and Transplant Surgery, University Hospital, Ludwig Maximilians University, Munich, Germany
| | - Felix Popp
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Florian Klein
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Laboratory of Experimental Immunology, Faculty of Medicine and University Hospital Cologne, Institute of Virology, University of Cologne Cologne, Germany
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, Cologne, Germany
| | - Michael S von Bergwelt-Baildon
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Department of Internal Medicine III, University Hospital, Ludwig Maximilians University, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Christiane J Bruns
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Kerstin Wennhold
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Hans A Schlößer
- Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| |
Collapse
|
27
|
Elfving H, Yu H, Fessehatsion KK, Brunnström H, Botling J, Gulyas M, Backman M, Lindberg A, Strell C, Micke P. Spatial distribution of tertiary lymphoid structures in the molecular and clinical context of non-small cell lung cancer. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01052-x. [PMID: 40029549 DOI: 10.1007/s13402-025-01052-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 03/05/2025] Open
Abstract
INTRODUCTION Tertiary lymphoid structures (TLS) are lymphocyte aggregates resembling secondary lymphoid organs and are pivotal in cancer immunity. The ambiguous morphological definition of TLS makes it challenging to ascertain their clinical impact on patient survival and response to immunotherapy. OBJECTIVES This study aimed to characterize TLS in hematoxylin-eosin tissue sections from lung cancer patients, assessing their occurrence in relation to the local immune environment, mutational background, and patient outcome. METHODS Two pathologists evaluated one whole tissue section from resection specimens of 680 NSCLC patients. TLS were spatially quantified within the tumor area or periphery and further categorized based on the presence of germinal centers (mature TLS). Metrics were integrated with immune cell counts, genomic and transcriptomic data, and correlated with clinical parameters. RESULTS TLS were present in 86% of 536 evaluable cases, predominantly in the tumor periphery, with a median of eight TLS per case. Mature TLS were found in 24% of cases. TLS presence correlated positively with increased plasma cell (CD138+) and lymphocytic cell (CD3+, CD8+, FOXP3+) infiltration. Tumors with higher tumor mutational burden exhibited higher numbers of peripheral TLS. The overall TLS quantity was independently associated with improved patient survival, irrespective of TLS maturation status. This prognostic association held true for peripheral TLS but not for tumor TLS. CONCLUSION TLS in NSCLC is common and their correlation with a specific immune phenotype suggests biological relevance in the local immune reaction. The prognostic significance of this scoring system on routine hematoxylin-eosin sections has the potential to augment diagnostic algorithms for NSCLC patients.
Collapse
Affiliation(s)
- Hedvig Elfving
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden.
| | - Hui Yu
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | | | - Hans Brunnström
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Johan Botling
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Miklos Gulyas
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Max Backman
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Amanda Lindberg
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Carina Strell
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Patrick Micke
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| |
Collapse
|
28
|
Fernandez-Cuesta L, Alcala N, Mathian E, Derks J, Thirlwell C, Dayton T, Marinoni I, Perren A, Walter T, Foll M. Basic science and translational implications of current knowledge on neuroendocrine tumors. J Clin Invest 2025; 135:e186702. [PMID: 40026252 DOI: 10.1172/jci186702] [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: 03/05/2025] Open
Abstract
Neuroendocrine tumors (NETs) are a diverse group of malignancies that can occur in various organs, with a notable prevalence in the lungs and gastrointestinal tract, which are the focus of this Review. Although NETs are rare in individual organs, their incidence has increased over recent decades, highlighting the urgent need for current classification systems to evolve by incorporating recent advances in the understanding of NET biology. Several omics studies have revealed molecular subtypes, which, when integrated into existing classification frameworks, may provide more clinically relevant insights for patients with NETs. This Review examines recent progress in elucidating the biology of NETs, with a particular emphasis on the tumor microenvironment and cells of origin. The existence of different cells of origin, which may contribute to distinct molecular groups, along with profiles of immune infiltration - despite being generally low - could explain the emergence of more aggressive cases and the potential for metastatic progression. Given the molecular heterogeneity of NETs and the diversity of their microenvironments and different cells of origin, there is an urgent need to develop morphomolecular classification systems. Such systems would make it possible to better characterize tumor progression, identify new therapeutic targets, and, ultimately, guide the development of personalized therapies.
Collapse
Affiliation(s)
- Lynnette Fernandez-Cuesta
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Nicolas Alcala
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Emilie Mathian
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Jules Derks
- Department of Pulmonary Medicine, Erasmus MC Cancer institute, University Medical Center, Rotterdam, Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Talya Dayton
- European Molecular Biology Laboratory Barcelona, Tissue Biology and Disease Modeling, Barcelona, Spain
| | - Ilaria Marinoni
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Aurel Perren
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Thomas Walter
- Service d'Oncologie Médicale, Groupement Hospitalier Centre, Institut de Cancérologie des Hospices Civils de Lyon, Lyon, France
| | - Matthieu Foll
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| |
Collapse
|
29
|
Westwood AC, Wilson BI, Laye J, Grabsch HI, Mueller W, Magee DR, Quirke P, West NP. Deep-learning enabled combined measurement of tumour cell density and tumour infiltrating lymphocyte density as a prognostic biomarker in colorectal cancer. BJC REPORTS 2025; 3:12. [PMID: 40033106 DOI: 10.1038/s44276-025-00123-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/20/2024] [Accepted: 01/17/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Within the colorectal cancer (CRC) tumour microenvironment, tumour infiltrating lymphocytes (TILs) and tumour cell density (TCD) are recognised prognostic markers. Measurement of TILs and TCD using deep-learning (DL) on haematoxylin and eosin (HE) whole slide images (WSIs) could aid management. METHODS HE WSIs from the primary tumours of 127 CRC patients were included. DL was used to quantify TILs across different regions of the tumour and TCD at the luminal surface. The relationship between TILs, TCD, and cancer-specific survival was analysed. RESULTS Median TIL density was higher at the invasive margin than the luminal surface (963 vs 795 TILs/mm2, P = 0.010). TILs and TCD were independently prognostic in multivariate analyses (HR 4.28, 95% CI 1.87-11.71, P = 0.004; HR 2.72, 95% CI 1.19-6.17, P = 0.017, respectively). Patients with both low TCD and low TILs had the poorest survival (HR 10.0, 95% CI 2.51-39.78, P = 0.001), when compared to those with a high TCD and TILs score. CONCLUSIONS DL derived TIL and TCD score were independently prognostic in CRC. Patients with low TILs and TCD are at the highest risk of cancer-specific death. DL quantification of TILs and TCD could be used in combination alongside other validated prognostic biomarkers in routine clinical practice.
Collapse
Affiliation(s)
- Alice C Westwood
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | | | - Jon Laye
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Heike I Grabsch
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, Netherlands
| | | | | | - Phillip Quirke
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Nicholas P West
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.
| |
Collapse
|
30
|
Lopez de Rodas M, Villalba-Esparza M, Sanmamed MF, Chen L, Rimm DL, Schalper KA. Biological and clinical significance of tumour-infiltrating lymphocytes in the era of immunotherapy: a multidimensional approach. Nat Rev Clin Oncol 2025; 22:163-181. [PMID: 39820025 DOI: 10.1038/s41571-024-00984-x] [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: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Immune-checkpoint inhibitors (ICIs) have improved clinical outcomes across several solid tumour types. Prominent efforts have focused on understanding the anticancer mechanisms of these agents, identifying biomarkers of response and uncovering resistance mechanisms to develop new immunotherapeutic approaches. This research has underscored the crucial roles of the tumour microenvironment and, particularly, tumour-infiltrating lymphocytes (TILs) in immune-mediated tumour elimination. Numerous studies have evaluated the prognostic and predictive value of TILs and the mechanisms that govern T cell dysfunction, fuelled by technical developments in single-cell transcriptomics, proteomics, high-dimensional spatial platforms and advanced computational models. However, questions remain regarding the definition of TILs, optimal strategies to study them, specific roles of different TIL subpopulations and their clinical implications in different treatment contexts. Additionally, most studies have focused on the abundance of major TIL subpopulations but have not developed standardized quantification strategies or analysed other crucial aspects such as their functional profile, spatial distribution and/or arrangement, tumour antigen-reactivity, clonal diversity and heterogeneity. In this Review, we discuss a conceptual framework for the systematic study of TILs and summarize the evidence regarding their biological properties and biomarker potential for ICI therapy. We also highlight opportunities, challenges and strategies to support future developments in this field.
Collapse
Affiliation(s)
- Miguel Lopez de Rodas
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Maria Villalba-Esparza
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Miguel F Sanmamed
- Department of Immunology and Immunotherapy, Centro de Investigación Médica Aplicada and Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - David L Rimm
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kurt A Schalper
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
31
|
Klein J, Tran WT, Viswanathan S, Salgado R, Poortmans P, Machiels M. Tumour-infiltrating Lymphocytes and Radiation Therapy in Rectal Cancer: Systematic Review and Meta-analysis. Clin Oncol (R Coll Radiol) 2025; 39:103742. [PMID: 39854781 DOI: 10.1016/j.clon.2024.103742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025]
Abstract
AIM Tumour-infiltrating lymphocytes (TILs) represent a promising cancer biomarker. Different TILs, including CD8+, CD4+, CD3+, and FOXP3+, have been associated with clinical outcomes. However, data are lacking regarding the value of TILs for patients receiving radiation therapy (RT). We conducted a systemic review and meta-analysis of available data evaluating TILs for patients receiving curative-intent therapy including RT. MATERIALS AND METHODS Eligible studies presented a defined cohort of patients who all received curative-intent therapy, including RT, and also reported the relationship between any TIL score and either tumour response or survival outcomes. After comprehensive search of online databases (PubMed, EMBASE, Cochrane, and Web of Science), 2 authors conducted title, abstract, and whole-text review for quality and risk of bias following Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) guidelines. Data from publications that met quality criteria were grouped via (1) TIL analysed, (2) pre- or post-RT TIL assessment, and (3) clinical outcome measured. RESULTS Initial search yielded 669 unique studies. Thirty-one studies met quality criteria, of which 20 studied rectal cancer (RC), 4 oesophageal, 3 pancreas, 2 lung, cervical/uterine 1 each. We conducted systematic review and meta-analysis of the RC publications. All except 2 were single-institutional cohort studies. After meta-analysis, the pre-RT epithelial CD8+ (p = 0.04) and stromal FOXP3+ (p = 0.01) counts were associated with survival without disease, while pre-RT epithelial (p = 0.02) and stromal (p = 0.001) FOXP3+ TILs were associated with overall survival. On post-RT analysis, epithelial (p = .04) and stromal (p = 0.02) CD8+ TILs were associated with survival without disease and epithelial CD8+ TILs were associated with overall survival (p = 0.01).Preoperative CD8+ and FOXP3+ TILs were generally associated with tumour response to RT, but meta-analysis was not conducted due to heterogeneity of response measurement techniques. CONCLUSION TILs represent a useful parameter for tumour response and survival outcomes for patients receiving curative-intent therapy, including RT for RC. Future work should aim to standardise TIL measurement and quantification methods and to develop protocols to clarify clinical application of these findings.
Collapse
Affiliation(s)
- J Klein
- Department of Radiation Oncology, State University of New York (SUNY) Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY, USA; Department of Radiation Oncology, Maimonides Medical Center, 6300 8th Ave, Brooklyn, NY, USA.
| | - W T Tran
- Department of Radiation Oncology, University of Toronto, 149 College St#504, Toronto, Ontario, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, Canada.
| | - S Viswanathan
- Department of Epidemiology and Population Health, Montefiore Einstein Comprehensive Cancer Center, 1300 Morris Park Ave, Block Building Room 315, Bronx, NY, USA.
| | - R Salgado
- Division of Research, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Australia; Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium.
| | - P Poortmans
- Faculty of Medicine and Health Sciences, University of Antwerp, Prinsstraat 13, 2000, Antwerp, Belgium; Department of Radiation Oncology, Iridium Netwerk, Oosterveldlaan 22, 2610, Antwerp, Belgium.
| | - M Machiels
- Faculty of Medicine and Health Sciences, University of Antwerp, Prinsstraat 13, 2000, Antwerp, Belgium; Department of Radiation Oncology, Iridium Netwerk, Oosterveldlaan 22, 2610, Antwerp, Belgium.
| |
Collapse
|
32
|
Ulkucu A, Erkaya M, Inal E, Gorgun E. The critical role of tumor size in predicting lymph node metastasis in early-stage colorectal cancer. Am J Surg 2025; 241:116152. [PMID: 39729965 DOI: 10.1016/j.amjsurg.2024.116152] [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: 07/30/2024] [Revised: 11/27/2024] [Accepted: 12/16/2024] [Indexed: 12/29/2024]
Abstract
BACKGROUND Main purpose of this study is to investigate impact of tumor size on risk of lymph node metastasis (LNM) in pT1-stage colorectal cancer (CRC), focusing on colon, rectosigmoid junction, and rectum. METHOD Patients diagnosed with primary pT1 CRC between 2015 and 2019 were selected from National Cancer Database, utilizing International Classification of Diseases for Oncology, Third Edition (ICD-O-3) codes. We analyzed factors influencing LNM using uni- and multivariate analysis, then isolated tumor size to study its impact on LNM. RESULTS In this study of 27,649 pT1-stage tumor patients, we found that 10 % of colon, 16 % of rectosigmoid junction, and 13 % of rectum were LNM+. The study had 14,339 males (51.97 %). Mean age was 64.9 (±11.7). In multivariate analysis, sample was adjusted by excluding confounding factors, isolating impact of tumor size on LNM. Analysis for only tumor size, patients with colon tumors >45 mm had 53 % increased odds of LNM (95 % CI [1.06, 2.23], p = 0.03), whereas tumor size did not significantly affect LNM in rectosigmoid and rectum cases, with odds ratios of 2.05 (95 % CI [0.82, 5.09], p = 0.12) and 1.62 (95 % CI [0.97, 2.71], p = 0.065) respectively, for tumors ≥45 mm compared to those <15 mm. CONCLUSION This investigation refines predictors of LNM, crucial for tailoring organ-sparing strategies in early-stage CRC management. While tumor size is significant determinant of LNM in colon cancer, early rectal and rectosigmoid cancers may be associated with lower risk of LNM.
Collapse
Affiliation(s)
- Attila Ulkucu
- Department of Colorectal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Metincan Erkaya
- Department of Colorectal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ekin Inal
- Department of Colorectal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Emre Gorgun
- Department of Colorectal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
33
|
Maggi E, Landolina N, Munari E, Mariotti FR, Tumino N, Vacca P, Azzarone B, Moretta L. T cells in the microenvironment of solid pediatric tumors: the case of neuroblastoma. Front Immunol 2025; 16:1544137. [PMID: 40092980 PMCID: PMC11906424 DOI: 10.3389/fimmu.2025.1544137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Neuroblastoma (NB) is an immunologically "cold" tumor with poor or no inflamed substrates as most of solid pediatric tumors (SPT). Consistent data indicate that NB tumor microenvironment (TME) is dominated by myeloid cells, with little (but variable) T cell infiltration. The obstacles to lymphocyte infiltration and to their anti-tumor activity are due to different tumor immune evasion strategies, including loss of HLA Class I molecules, high expression of immune checkpoint molecular ligands leading to exhaustion of T effector (and NK) cells, induction of T regulatory, myeloid and stromal cells and secretion of immunosuppressive mediators. In odds with adult solid tumors, NB displays weak immunogenicity caused by intrinsic low mutational burden and scant expression of neoepitopes in the context of MHC-class I antigens which, in turn, are particularly poorly expressed on NB cells, thus inducing low anti-tumor T cell responses. In addition, NB is generated from embryonal cells and is the result of transcriptional abnormalities and not of the accumulation of genetic mutations over time, thus further explaining the low immunogenicity. The poor expression of immunogenic molecules on tumor cells is associated with the high production of immunosuppressive factors which further downregulate lymphocyte infiltration and activity, thus explaining the limited efficacy of new drugs in NB, as immune checkpoint inhibitors. This review is focused on examining the role of T effector and regulatory cells infiltrating TME of NB, taking into account their repertoire, phenotype, function, plasticity and, importantly, predictive value for defining novel targets for therapy.
Collapse
Affiliation(s)
- Enrico Maggi
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nadine Landolina
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Enrico Munari
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | | | - Nicola Tumino
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paola Vacca
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Bruno Azzarone
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
34
|
Nagy MZ, Plaza-Rojas LB, Boucher JC, Kostenko E, Austin AL, Tarhini AA, Chen Z, Du D, Ojwang' AME, Davis J, Obermayer A, Rejniak KA, Shaw TI, Guevara-Patino JA. Effector T cells under hypoxia have an altered transcriptome similar to tumor-stressed T cells found in non-responsive melanoma patients. J Immunother Cancer 2025; 13:e010153. [PMID: 40010774 DOI: 10.1136/jitc-2024-010153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND In the tumor microenvironment (TME), hypoxia stands as a significant factor that modulates immune responses, especially those driven by T cells. As T cell-based therapies often fail to work in solid tumors, this study aims to investigate the effects of hypoxia on T cell topo-distribution in the TME, gene expression association with T cell states, and clinical responses in melanoma. METHODS To generate detailed information on tumor oxygenation and T cell accessibility, we used mathematical modeling of human melanoma tissue microarrays that incorporate oxygen supply from vessels, intratumoral diffusion, and cellular uptake. We created tumor maps and derived plots showing the fraction of CD4 and CD8 T cells against the distance to the nearest vessel and oxygen pressure. To assess their function and transcriptional changes caused by hypoxia, effector T cells were generated and cultured under hypoxia (0.5% oxygen) or normoxia (21% oxygen). The T cell hypoxia-transcriptional signature was compared against datasets from msigDB, iATLAS (clinical trials of melanoma patients treated with immune checkpoint inhibitors (ICIs)), ORIEN AVATAR (real-world melanoma patients treated with ICIs), and a single-cell atlas of tumor-infiltrating lymphocytes. RESULTS We made three specific observations: (1) in melanoma T cells preferentially accumulated in oxygenated areas close to blood vessels (50-100 µm from the vasculature in the regions of high oxygen availability) but not in hypoxic areas far from blood vessels. (2) Our analysis confirmed that under hypoxia, T cell functions were significantly reduced compared with normoxic conditions and accompanied by a unique gene signature. Furthermore, this hypoxic gene signature was prevalent in resting and non-activated T cells. Notably and clinically relevant, the hypoxic T cell gene set was found to correlate with reduced overall survival and reduced progression-free survival in melanoma patients, which was more pronounced in non-responder patients undergoing ICI therapy. (3) Finally, compared with a single-cell atlas of tumor-infiltrating T cells, our hypoxia signature aligned with a population of cells at a state termed stress response state (TSTR). CONCLUSIONS Our study highlights the critical role of hypoxia in shaping T cell distribution and its correlation with clinical outcomes in melanoma. We revealed a preferential accumulation of T cells in oxygenated areas. Moreover, hypoxic T cells develop a distinct hypoxic gene signature prevalent in resting, non-activated T cells and TSTR that was also associated with poorer outcomes, particularly pronounced among non-responders to ICIs.
Collapse
Affiliation(s)
- Mate Z Nagy
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Lourdes B Plaza-Rojas
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Justin C Boucher
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Elena Kostenko
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Anna L Austin
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Ahmad A Tarhini
- Departments of Cutaneous Oncology and Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Zhihua Chen
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Dongliang Du
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Awino Maureiq E Ojwang'
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Joshua Davis
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Alyssa Obermayer
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Katarzyna A Rejniak
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Timothy I Shaw
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Jose A Guevara-Patino
- Department of Immunology, H Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| |
Collapse
|
35
|
Dzhalilova D, Silina M, Kosyreva A, Fokichev N, Makarova O. Morphofunctional changes in the immune system in colitis-associated colorectal cancer in tolerant and susceptible to hypoxia mice. PeerJ 2025; 13:e19024. [PMID: 40028198 PMCID: PMC11869898 DOI: 10.7717/peerj.19024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Background One of the effective strategies for the treatment of tumor diseases, including colitis-associated colorectal cancer (CAC), is immunotherapy. During inflammation, NF-κB is activated, which is connected with the hypoxia-inducible factor-HIF, regulating the immune cells functioning and influences the CAC development. Organisms differ according to their hypoxia resistance and HIF expression. Therefore, the aim of the study was to characterize the thymus, spleen and mesenteric lymph nodes morphofunctional features, as well as changes in the subpopulation composition of peripheral blood cells and mesenteric lymph nodes in tolerant and susceptible to hypoxia C57Bl/6 mice in CAC. Methods Hypoxia tolerance was assessed by gasping time measurement in hypobaric decompression chamber. Based on the outcome, the mice were assigned to three groups characterized as 'tolerant to hypoxia', 'normal', and 'susceptible to hypoxia'. A month after determining hypoxia resistance CAC was modeled by intraperitoneal azoxymethane (AOM) administration and three cycles of dextran sulfate sodium consumption. Mice were sacrificed on the 141st day after the AOM administration, a morphological, morphometric and immunohistochemical study of tumors, morphological and morphometric study of thymus and spleen, and subpopulation composition of peripheral blood cells and mesenteric lymph nodes assessment were carried out. Results Tumors in tolerant and susceptible to hypoxia mice were represented by glandular intraepithelial neoplasia and adenocarcinomas, the area of which was larger in susceptible mice. Immunohistochemical study revealed a more pronounced Ki-67+ staining in tumors of susceptible mice. In CAC, only in tolerant mice, expansion of the thymic cortex was observed relative to the control group, while in susceptible ones, no changes were detected. Only in susceptible to hypoxia mice, spleen germinal centers of lymphoid follicles enlargement were observed. Only in susceptible mice during CAC, in comparison to the control group, the relative and absolute number of B-lymphocytes and relative-cytotoxic T-lymphocytes in blood increased. The relative cytotoxic T-lymphocytes and NK cells number in peripheral blood during CAC was higher in susceptible to hypoxia mice compared to tolerant ones. In susceptible to hypoxia mice, more pronounced changes in the mesenteric lymph nodes subpopulation composition of cells were revealed-only in them the absolute and relative number of B-lymphocytes and NK cells, the absolute number of cytotoxic T-lymphocytes increased, and the relative number of macrophages decreased. Conclusions Morphofunctional differences in the thymus, spleen, mesenteric lymph nodes and blood immune cells reactions indicated the more pronounced immune response to the CAC development in susceptible to hypoxia mice, which should be taken into account in experimental studies.
Collapse
Affiliation(s)
- Dzhuliia Dzhalilova
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - Maria Silina
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - Anna Kosyreva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, People’s Friendship University of Russia (RUDN University), Moscow, Russia
| | - Nikolai Fokichev
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia
| | - Olga Makarova
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
| |
Collapse
|
36
|
Sirniö P, Elomaa H, Tuomisto A, Äijälä VK, Karjalainen H, Kastinen M, Tapiainen VV, Sirkiä O, Ahtiainen M, Helminen O, Wirta EV, Rintala J, Meriläinen S, Saarnio J, Rautio T, Seppälä TT, Böhm J, Mecklin JP, Mäkinen MJ, Väyrynen JP. CDX2 and SATB2 loss are associated with myeloid cell infiltration and poor survival in colorectal cancer. Cancer Immunol Immunother 2025; 74:111. [PMID: 39998677 PMCID: PMC11861821 DOI: 10.1007/s00262-025-03964-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: 10/11/2024] [Accepted: 01/29/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Caudal-type homeobox 2 (CDX2) and special AT-rich sequence-binding protein 2 (SATB2) are transcription factors playing important roles in intestinal homeostasis and participating in the regulation of intestinal inflammation. In colorectal cancer (CRC), reduced expression levels of CDX2 and SATB2 have been associated with poor differentiation and worse survival. However, their prognostic significance still needs further clarification, and the associations between CDX2 and SATB2 and immune cell infiltration into the CRC microenvironment are largely unknown. METHODS We analyzed CDX2 and SATB2 expression in two large cohorts of stages I-IV CRC patients (N = 2302) and analyzed their associations with clinicopathologic parameters, the density of local immune cells (determined with three multiplex immunohistochemistry panels and conventional immunohistochemistry), and survival. RESULTS In mismatch repair-proficient tumors, reduced CDX2 and SATB2 expression were associated with higher densities of immature monocytic cells, macrophages, and M2-like macrophages. Low expression of CDX2 was associated with shorter cancer-specific survival independent of conventional prognostic parameters in both cohorts. In the larger cohort, adjusted hazard ratio (HR) for negative (vs. high) CDX2 expression was 3.62 (95% CI 2.08-6.31, ptrend < 0.0001), and adjusted HR for negative (vs. high) SATB2 level was 1.61 (95% CI 0.97-2.67, ptrend = 0.002). CONCLUSION This study indicates that reduced CDX2 and SATB2 expression levels are associated with myeloid cell infiltration in the CRC microenvironment and represent markers for poor outcome. These findings highlight the potential of CDX2 and SATB2 as biomarkers for classifying CRC patients and support their role in regulating the tumor microenvironment.
Collapse
Affiliation(s)
- Päivi Sirniö
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Hanna Elomaa
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Department of Education and Research, Well Being Services County of Central Finland, Jyväskylä, Finland
| | - Anne Tuomisto
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Ville K Äijälä
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Henna Karjalainen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Meeri Kastinen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Vilja V Tapiainen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Onni Sirkiä
- Department of Pathology, Hospital Nova of Central Finland, Well Being Services County of Central Finland, Jyväskylä, Finland
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Maarit Ahtiainen
- Department of Pathology, Hospital Nova of Central Finland, Well Being Services County of Central Finland, Jyväskylä, Finland
| | - Olli Helminen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Erkki-Ville Wirta
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
| | - Jukka Rintala
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Sanna Meriläinen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Juha Saarnio
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Tero Rautio
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Toni T Seppälä
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
- Department of Gastrointestinal Surgery, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics, Research Program Unit, University of Helsinki, Helsinki, Finland
| | - Jan Böhm
- Department of Pathology, Hospital Nova of Central Finland, Well Being Services County of Central Finland, Jyväskylä, Finland
| | - Jukka-Pekka Mecklin
- Department of Education and Research, Well Being Services County of Central Finland, Jyväskylä, Finland
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Markus J Mäkinen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland
| | - Juha P Väyrynen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Aapistie 5A, 90220, Oulu, Finland.
| |
Collapse
|
37
|
Shi J, Zhao L, Wang K, Lin J, Shen J. Disulfidptosis classification of pancreatic carcinoma reveals correlation with clinical prognosis and immune profile. Hereditas 2025; 162:26. [PMID: 39987145 PMCID: PMC11846472 DOI: 10.1186/s41065-025-00381-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/27/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Disulfidptosis, a novel form of metabolism-related regulated cell death, is a promising intervention for cancer therapeutic intervention. Although aberrant expression of long-chain noncoding RNAs (lncRNAs) expression has been associated with pancreatic carcinoma (PC) development, the biological properties and prognostic potential of disulfidptosis-related lncRNAs (DRLs) remain unclear. METHODS We obtained RNA-seq data, clinical data, and genomic mutations of PC from the TCGA database, and then determined DRLs. We developed a risk score model and analyzed the role of risk score in the predictive ability, immune cell infiltration, immunotherapy response, and drug sensitivity. RESULTS We finally established a prognostic model including three DRLs (AP005233.2, FAM83A-AS1, and TRAF3IP2-AS1). According to Kaplan-Meier curve analysis, the survival time of patients in the low-risk group was significantly longer than that in the high-risk group. Based on enrichment analysis, significant associations between metabolic processes and differentially expressed genes were assessed in two risk groups. In addition, we observed significant differences in the tumor immune microenvironment landscape. Tumor Immune Dysfunction and Rejection (TIDE) analysis showed no statistically significant likelihood of immune evasion in both risk groups. Patients exhibiting both high risk and high tumor mutation burden (TMB) had the poorest survival times, while those falling into the low risk and low TMB categories showed the best prognosis. Moreover, the risk group identified by the 3-DRLs profile showed significant drug sensitivity. CONCLUSIONS Our proposed 3-DRLs-based feature could serve as a promising tool for predicting the prognosis, immune landscape, and treatment response of PC patients, thus facilitating optimal clinical decision-making.
Collapse
Affiliation(s)
- Jiangmin Shi
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Liang Zhao
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Kai Wang
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Jieqiong Lin
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China
| | - Jianwei Shen
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to, Ningbo University), Ningbo, Zhejiang Province, 315040, P.R. China.
| |
Collapse
|
38
|
Lee SH, Pankaj A, Yilmaz O, Deshpande V, Yilmaz O. Beta-2-microglobulin positive tumor cells and CD8 positive lymphocytes are associated with outcome in post-neoadjuvant colorectal cancer resections. Hum Pathol 2025; 155:105737. [PMID: 39988058 DOI: 10.1016/j.humpath.2025.105737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Locally advanced colorectal cancers are treated with neoadjuvant therapy (NAT), which has been shown to alter the characteristics of the tumor including size, lymph node yield, and histologic grade. We seek to interrogate the effect of NAT on the immune microenvironment. We compared 190 patients with colorectal adenocarcinoma treated with NAT with those without NAT (n = 926). We evaluated clinicopathologic and molecular factors and performed immunohistochemistry and quantification on tissue microarrays for HLA class I/II proteins, beta-2-microglobulin (B2M), CD8, CD163, LAG3, PD-L1, and FoxP3. Patients in the NAT group were younger (60.9 vs 67.9, p < 0.001) and more often male (59.5 vs. 47.9, p = 0.004) than those in the non-NAT group. Tumors in the NAT group were smaller (3.5 vs 4.7 cm, p < 0.001), less often high grade (6.5% vs. 16.2%, p = 0.001), more frequently in the rectum (68.9% vs. 6.6%, p < 0.001) and associated with lower lymph node yields (p = 0.002); however, the incidence of extramural venous invasion, perineural invasion, and AJCC stage 3-4 disease were not different. Immune cells positive for CD8 (p = 0.011) were significantly lower in the NAT group. A high number of CD8+ cells and higher expression of B2M in tumor cells showed a significant survival benefit in both NAT and non-NAT group. NAT is associated with an immune-low tumor environment. CD8+ cells and tumor B2M expression may help identify a subset of immune high-tumors following NAT. This identification could aid in determining patients who may benefit from conservative management of colorectal carcinomas.
Collapse
Affiliation(s)
- Soo Hyun Lee
- Department of Pathology, Boston Medical Center, Boston, MA, USA
| | - Amaya Pankaj
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Omer Yilmaz
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Vikram Deshpande
- Harvard Medical School, Boston, MA, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Osman Yilmaz
- Harvard Medical School, Boston, MA, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
39
|
Vonica RC, Morgovan C, Butuca A, Pumnea M, Cipaian RC, Frum A, Dobrea CM, Vonica-Tincu AL, Pacnejer AM, Batar F, Vornicu V, Ghibu S, Gligor FG. Real-World Evidence of Bevacizumab and Panitumumab Drug Resistance and Drug Ineffectiveness from EudraVigilance Database. Cancers (Basel) 2025; 17:663. [PMID: 40002260 PMCID: PMC11853327 DOI: 10.3390/cancers17040663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/02/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world, with an average 5-year overall survival (OS) rate of approximately 60% [...].
Collapse
Affiliation(s)
- Razvan Constantin Vonica
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Claudiu Morgovan
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Anca Butuca
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Manuela Pumnea
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Remus Calin Cipaian
- Clinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania;
- County Clinical Emergency Hospital of Sibiu, 2-4 Corneliu Coposu Str., 550245 Sibiu, Romania
| | - Adina Frum
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Carmen Maximiliana Dobrea
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Andreea Loredana Vonica-Tincu
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Aliteia-Maria Pacnejer
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Florina Batar
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Vlad Vornicu
- Department IX Surgery, Discipline of Oncology, Faculty of Medicine, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania;
| | - Steliana Ghibu
- Department of Pharmacology, Physiology and Pathophysiology, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Felicia Gabriela Gligor
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| |
Collapse
|
40
|
Jiang M, Sun J, Hu C, Wu L, Fan Y, Wang Z, Liu L, Wu C, Wu F, Gao G, Li F, Wang L, Li X, Cheng L, Peng B, Zhou H, Zhou C. A tumor cornification and immune-infiltration-based scheme for anti-PD-1 plus chemotherapy response in advanced squamous cell lung carcinoma. MED 2025; 6:100516. [PMID: 39395411 DOI: 10.1016/j.medj.2024.09.005] [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: 12/26/2023] [Revised: 07/29/2024] [Accepted: 09/13/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Anti-PD-1 immunotherapy plus chemotherapy (combo) exhibits significantly prolonged survival for squamous cell lung cancer (LUSC). An exploration of predictive biomarkers is still needed. METHODS High-throughput RNA sequencing (RNA-seq) of 349 LUSC samples from the randomized, multi-center, phase 3 trial ORIENT-12 (ClinicalTrials.gov: NCT03629925) was conducted for biomarker discovery, followed by flow cytometry and multiplex immunohistochemistry (mIHC) in additional clinical cohorts, and in vitro experiments were performed for verification. RESULTS A high abundance of activated CD8+ T and CD56bright natural killer (NK) cells benefited patients' outcomes (progression-free survival [PFS]; overall survival [OS]) with combo treatment. Tumor cornification level remarkably affected the infiltration of the two crucial immune cells. Thus, a novel scheme of LUSC immune infiltration and cornification characterization-based classification (LICC) was established for combo efficacy prediction. Patients who received combo treatment achieved significant PFS improvements in LICC1 (hazard ratio [HR] = 0.43, 95% confidence interval [CI]: 0.25-0.75, p = 0.0029) and LICC2 (HR = 0.32, 95% CI: 0.17-0.58, p = 0.0002) subtypes but not in the LICC3 subtype (HR = 0.86, 95% CI: 0.60-1.23, p = 0.4053). Via single-cell RNA-seq analysis, the tumor cornification signal was mainly mapped to SPRR3+ tumor cells, whose relationships with activated CD8+ T or CD56bright NK cells were verified using flow cytometry and mIHC. Our data suggest that SPRR3+ tumor cells might evade immune surveillance via the CD24-SIGLEC10 (M2 macrophage) axis to maintain a suppressive tumor microenvironment. CONCLUSIONS Tumor cornification greatly impacts immune infiltration, and the LICC scheme may guide clinical medication of anti-PD-1+chemo treatment in patients with LUSC. FUNDING The study was funded by the National Key R&D Program of China, the National Natural Science Foundation of China, Shanghia Multidisplinary Cooperation Building Project for Diagnosis and Treatment of Major Disease, and Innovent Biologics, Inc.
Collapse
Affiliation(s)
- Minlin Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China; Medical School, Tongji University, Shanghai 200433, China
| | - Jiya Sun
- Innovent Biologics, Inc., Suzhou, Jiangsu 215123, China
| | - Congli Hu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China; Medical School, Tongji University, Shanghai 200433, China
| | - Lin Wu
- Thoracic Medicine Department II, Hunan Cancer Hospital, Changsha, Hunan 410031, China
| | - Yun Fan
- Oncology Department, Cancer Hospital of the University of Chinese Academy of Science, Hangzhou, Zhejiang 310005, China
| | - Zhehai Wang
- Respiratory Department, Shandong Cancer Hospital, Jinan, Shandong 250117, China
| | - Lianke Liu
- Oncology Department, Jiangsu Province Hospital, Nanjing, Jiangsu 210029, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai 200433, China
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Guanghui Gao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Fei Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Frontier Innovation Center, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Lei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Xuefei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Lei Cheng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Bo Peng
- Innovent Biologics, Inc., Suzhou, Jiangsu 215123, China
| | - Hui Zhou
- Innovent Biologics, Inc., Suzhou, Jiangsu 215123, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai East Hospital, Shanghai 200120, China.
| |
Collapse
|
41
|
Dumut DC, Hajduch M, Zacharias AM, Duan Q, Frydrych I, Rozankova Z, Popper M, Garic D, Paun RA, Centorame A, Shah J, Mistrik M, Dzubak P, De Sanctis JB, Radzioch D. Diethyldithiocarbamate-copper complex ignites the tumor microenvironment through NKG2D-NKG2DL axis. Front Immunol 2025; 16:1491450. [PMID: 40013140 PMCID: PMC11860975 DOI: 10.3389/fimmu.2025.1491450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/17/2025] [Indexed: 02/28/2025] Open
Abstract
Advanced metastatic colorectal cancer (CRC) with deficient DNA mismatch repair (MMR-d), or immune-hot CRCs, show significantly improved clinical outcomes compared to MMR-proficient (MMR-p), or immune-cold CRCs. While the prior represents about 5% of all CRCs, the latter represent 95% and are characterized by low immunogenicity. This study investigates bis-diethyldithiocarbamate (CuET), a novel anticancer compound, and its impact on the colorectal cancer tumor microenvironment (TME). CuET is shown to convert immunologically inactive tumors into hotbeds of antitumor immune responses, marked by increased lymphocyte infiltration, heightened cytotoxicity of natural killer (NK) and T cells, and enhanced non-self recognition by lymphocytes. The potent anticancer cytotoxicity and in vivo safety and efficacy of CuET are established. In summary, CuET transforms the colorectal cancer TME, bolstering NK and T cell cytotoxicity and refining tumor cell recognition through non-classical activation via the NKG2D/NKG2DL axis. This study unveils a novel mechanism of action for CuET: a potent immunomodulator capable of turning cold tumors hot.
Collapse
Affiliation(s)
- Daciana C. Dumut
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Amanda M. Zacharias
- Department of Biomedical & Molecular Sciences, Faculty of Health Sciences, Queen’s University, Kingston, ON, Canada
| | - Qingling Duan
- Department of Biomedical & Molecular Sciences, Faculty of Health Sciences, Queen’s University, Kingston, ON, Canada
- School of Computing, Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Ivo Frydrych
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - Zuzana Rozankova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Miroslav Popper
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Dusan Garic
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Radu Alexandru Paun
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Amanda Centorame
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
| | - Juhi Shah
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
| | - Martin Mistrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Petr Dzubak
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Juan B. De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Danuta Radzioch
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| |
Collapse
|
42
|
De Lucia A, Mazzotti L, Gaimari A, Zurlo M, Maltoni R, Cerchione C, Bravaccini S, Delmonte A, Crinò L, Borges de Souza P, Pasini L, Nicolini F, Bianchi F, Juan M, Calderon H, Magnoni C, Gazzola L, Ulivi P, Mazza M. Non-small cell lung cancer and the tumor microenvironment: making headway from targeted therapies to advanced immunotherapy. Front Immunol 2025; 16:1515748. [PMID: 39995659 PMCID: PMC11847692 DOI: 10.3389/fimmu.2025.1515748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
Over the past decades, significant progress has been made in the understanding of non-small cell lung cancer (NSCLC) biology and tumor progression mechanisms, resulting in the development of novel strategies for early detection and wide-ranging care approaches. Since their introduction, over 20 years ago, targeted therapies with tyrosine kinase inhibitors (TKIs) have revolutionized the treatment landscape for NSCLC. Nowadays, targeted therapies remain the gold standard for many patients, but still they suffer from many adverse effects, including unexpected toxicity and intrinsic acquired resistance mutations, which lead to relapse. The adoption of immune checkpoint inhibitors (ICIs) in 2015, has offered exceptional survival benefits for patients without targetable alterations. Despite this notable progress, challenges remain, as not all patients respond favorably to ICIs, and resistance to therapy can develop over time. A crucial factor influencing clinical response to immunotherapy is the tumor microenvironment (TME). The TME is pivotal in orchestrating the interactions between neoplastic cells and the immune system, influencing tumor growth and treatment outcomes. In this review, we discuss how the understanding of this intricate relationship is crucial for the success of immunotherapy and survey the current state of immunotherapy intervention, with a focus on forthcoming and promising chimeric antigen receptor (CAR) T cell therapies in NSCLC. The TME sets major obstacles for CAR-T therapies, creating conditions that suppress the immune response, inducing T cell exhaustion. To enhance treatment efficacy, specific efforts associated with CAR-T cell therapy in NSCLC, should definitely focus TME-related immunosuppression and antigen escape mechanisms, by combining CAR-T cells with immune checkpoint blockades.
Collapse
Affiliation(s)
- Anna De Lucia
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucia Mazzotti
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Anna Gaimari
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Matteo Zurlo
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Roberta Maltoni
- Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sara Bravaccini
- Department of Medicine and Surgery, “Kore” University of Enna, Enna, Italy
| | - Angelo Delmonte
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucio Crinò
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Patricia Borges de Souza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Luigi Pasini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabio Nicolini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabrizio Bianchi
- Unit of Cancer Biomarker, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Manel Juan
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Hugo Calderon
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Chiara Magnoni
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luca Gazzola
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paola Ulivi
- Translational Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Massimiliano Mazza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
43
|
Huang A, Wu J, Wang J, Jiao C, Yang Y, Xiao H, Yao L. Immune gene features and prognosis in colorectal cancer: insights from ssGSEA typing. Discov Oncol 2025; 16:139. [PMID: 39921789 PMCID: PMC11807041 DOI: 10.1007/s12672-025-01928-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/04/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a molecularly heterogeneous disease, and its treatment and prognosis vary greatly among subgroups. Therefore, it is necessary to identify prognostic factors associated with the biological heterogeneity of CRC in order to improve patients' survival expectations. METHODS We obtained and merged RNA-Seq data along with clinical details for colorectal cancer (CRC) from The Cancer Genome Atlas (TCGA) repository, and then performed immunocluster typing on all CRC specimens. We conducted differential expression gene (DEG) analysis, gene set enrichment analysis (GSEA), and tumor microenvironment (TME) analysis on CRC samples that were divided into high and low Immunity categories. Moreover, we pinpointed prognostic genes from immune-related gene (IRGs) sets, developed a prognostic risk model, and executed survival analysis, receiver operating characteristic (ROC) curve analysis, and independent prognostic analysis. Additionally, we assessed the risk for patients categorized into high- and low-risk groups based on the model. Lastly, we created a Nomogram to customize the prediction of survival outcomes in CRC patients. RESULTS CRC samples were divided into high and low Immunity groups based on the median value of the immunity score. Between the two groups, a total of 1550 DEGs were identified and 395 differentially expressed immune-related genes (DE-IRGs) were identified by intersection with 2483 IRGs. The DE-IRGs of the high Immunity group were dominated by Cytokine receptor interactions, chemokine signaling pathways and immune cell-mediated cytotoxicity, and molecule function of immune effector process. TME analysis showed that most of the 27 immune cells and functions were highly enriched in high Immunity group, whose Immune Score, Stromal Score and ESTIMATE Score were significantly higher. Subsequently, a prognostic risk model of CRC was constructed based on 12 prognostic genes, and the accuracy and reliability of the model prediction were verified. Finally, Nomogram enabled accurate individual prediction of the survival prognosis of CRC patients. CONCLUSIONS Our study develops an immune-related prognostic model and Nomogram that reliably predicts survival outcomes in CRC patients and enhances understanding of the tumor immunity and molecular mechanisms of CRC.
Collapse
Affiliation(s)
- Anwen Huang
- Department of Hepatopancreatobiliary Surgery, Shanghai Punan Hospital, Shanghai, 200125, China
| | - Jinxiu Wu
- Department of General Surgery, Shanghai Punan Hospital, Shanghai, 200125, China
| | - Jiakuan Wang
- Department of General Surgery, Shanghai Punan Hospital, Shanghai, 200125, China
| | - Chengwen Jiao
- Department of General Surgery, Shanghai Punan Hospital, Shanghai, 200125, China
| | - Yunfei Yang
- Department of General Surgery, Shanghai Punan Hospital, Shanghai, 200125, China
| | - Huaiwen Xiao
- Department of General Surgery, Shanghai Punan Hospital, Shanghai, 200125, China
| | - Li Yao
- Department of General Surgery, Shanghai Punan Hospital, Shanghai, 200125, China.
| |
Collapse
|
44
|
Abrantes AM, Caetano-Oliveira R, Oliveiros B, Cipriano MA, Tralhão JG. Association Between Colorectal Cancer Primary Features and Liver Metastases Histological Growth Patterns: Inflammation on the Primary Tumor is Associated with Desmoplastic Growth Pattern. Clin Colorectal Cancer 2025:S1533-0028(25)00016-7. [PMID: 40021416 DOI: 10.1016/j.clcc.2025.01.004] [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: 06/03/2024] [Revised: 01/04/2025] [Accepted: 01/29/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND More than 50% of patients diagnosed with colorectal cancer (CRC) will develop liver metastases (CRCLM), which is the main cause of death for more than 60% of these patients. The aim of this study was to correlate the clinical and pathological characteristics of the primary CRC and CRCLM, with emphasis in predicting the histological growth pattern of the CRCLM. METHODS Cohort of 73 patients with CRC. Analysis of clinical data and blinded pathological review was performed related with primary tumor and CRCLM features. The analysis was performed in SPSS (version 27) with a significance level of 5%. RESULTS A statistically significant association was found between tumor size and metastasis growth pattern (P = .002), with larger tumors giving rise to metastases with a nondesmoplastic growth pattern. Lymphovascular invasion (LVI) was associated with metachronous CRCLM (P = .043). In the absence of LVI, the time required for CRCLM to appear was significantly longer (P = .011). The number of metastases was significantly higher (P = .049) in tumors without LVI when compared to tumors with LVI. There was a statistically significant association between CRC high-grade inflammation and the desmoplastic metastases growth pattern of the CRCLM (P = .017). CONCLUSION The possibility of predicting the CRCLM histological growth pattern resorting to primary CRC characteristics would be useful for proper patient selection for surgery and adapting biological therapies.
Collapse
Affiliation(s)
- Ana Margarida Abrantes
- Biophysics Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal; Coimbra Centor Académico e Clínico (CACC), Coimbra, Portugal
| | - Rui Caetano-Oliveira
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal; Coimbra Centor Académico e Clínico (CACC), Coimbra, Portugal; Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; Centro de Anatomia Patológica Germano de Sousa, Coimbra, Portugal; General Surgery Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.
| | - Bárbara Oliveiros
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal; Coimbra Centor Académico e Clínico (CACC), Coimbra, Portugal
| | | | - José Guilherme Tralhão
- Biophysics Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal; Coimbra Centor Académico e Clínico (CACC), Coimbra, Portugal; General Surgery Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| |
Collapse
|
45
|
Kar S, Verma D, Mehrotra S, Prajapati VK. Reconfiguring the immune system to target cancer: Therapies based on T cells, cytokines, and vaccines. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:77-150. [PMID: 39978976 DOI: 10.1016/bs.apcsb.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Over the years, extensive research has been dedicated to performing in-depth analysis of cancer to uncover the intricate details of its nature - including the types of cancer, causative agents, stimulators of disease progression, factors contributing to poor prognosis, and efficient therapies to restrict the metastatic aggressiveness. This chapter highlights the mechanisms through which different arms of the host immune system - namely cytokines, lymphocytes, antigen-presenting cells (APCs) -can be mobilized to eradicate cancer. Most malignant tumors are either poorly immunogenic, or are harbored in a highly immuno-suppressive microenvironment. This is why reinforcing the host's anti-tumor defenses, through infusion of pro-inflammatory cytokines, tumor antigen-loaded APCs, and anti-tumor cytotoxic cells has emerged as a viable treatment option against cancer. The chapter also highlights the ongoing preclinical and clinical studies in different malignancies and the outcome of various therapies. Although these methods are not foolproof, and antigen escape variants can still evade or develop resistance to customized therapies, they achieve disease stabilization in several cases when conventional treatments fail. In many instances, combination therapies involving cytokines, T cells, and vaccinations prove more effective than monotherapies. The limitations of the current therapies are also discussed, along with ongoing modifications aimed at improving efficacy.
Collapse
Affiliation(s)
- Sramona Kar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Divya Verma
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
46
|
Liao K, Zhu M, Guo L, Gao Z, Cheng J, Sun B, Qian Y, Lin B, Zhang J, Qian T, Jiang Y, Xu Y, Zhong Q, Wang X. Assessment of prognosis and responsiveness to immunotherapy in colorectal cancer patients based on the level of immune cell infiltration. Front Immunol 2025; 16:1514238. [PMID: 39963131 PMCID: PMC11830669 DOI: 10.3389/fimmu.2025.1514238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Abstract
Objective To build a new prognostic risk assessment model based on immune cell co-expression networks for predicting overall survival and evaluating the efficacy of immunotherapy for colon cancer patients. Methods The Cancer Genome Atlas (TCGA) database was used to obtain mRNA expression profiling data, clinical information, and somatic mutation data from colorectal cancer patients. The degree of tumor immune cell infiltration of the samples was analyzed using the CIBERSORT algorithm. Co-expression of immune-related genes was analyzed using weighted correlation network analysis (WGCNA) and gene modules were identified. Prognosis-related genes were screened and models were constructed using LASSO-Cox analysis. The models were validated by survival analysis. The prognostic potential of the models was quantitatively assessed using Cox regression analysis and the development of column line plots. Immunotherapy sensitivity analysis was performed using CIBERSORT and TIMER algorithms. Gene biofunction analysis was performed using Gene set enrichment analysis (GSEA) and Gene set variation analysis (GSVA). And the chemotherapeutic response to different drugs was assessed. Results We established a novel prognostic model utilizing the WGCNA method, which demonstrated robust predictive accuracy for patient survival. The high-risk subgroup in our model exhibited elevated immune cell infiltration coupled with a higher tumor mutation burden, but the difference in response to immunotherapy was not significant compared to the low-risk group. Furthermore, we identified distinct chemotherapy responses to 39 drugs between these risk subgroups. Conclusion This study revealed a significant correlation between high levels of immune infiltration and unfavorable prognosis in patients with colon cancer. Furthermore, an accurate prognostic risk prediction model based on the co-expression of relevant genes by immune cells was developed, enabling precise prediction of survival of colon cancer patients. These findings offer valuable insights for accurate prognostication and comprehensive management of individuals diagnosed with colon cancer.
Collapse
Affiliation(s)
- Kaili Liao
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Minqi Zhu
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Lei Guo
- The 2 Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zijun Gao
- The 2 Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jinting Cheng
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bing Sun
- Queen Mary College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yihui Qian
- The 2 Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bingying Lin
- Queen Mary College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jingyan Zhang
- Queen Mary College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Tingyi Qian
- The 1 Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yixin Jiang
- Queen Mary College, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yanmei Xu
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qionghui Zhong
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
47
|
Gorría T, Sierra-Boada M, Rojas M, Figueras C, Marin S, Madurga S, Cascante M, Maurel J. Metabolic Singularities in Microsatellite-Stable Colorectal Cancer: Identifying Key Players in Immunosuppression to Improve the Immunotherapy Response. Cancers (Basel) 2025; 17:498. [PMID: 39941865 PMCID: PMC11815897 DOI: 10.3390/cancers17030498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Although immune checkpoint inhibitor (ICI) therapy is currently the standard of care in microsatellite-unstable (MSI) metastatic colorectal cancer (CRC), ICI therapy, alone or in combination with other therapies, is not a treatment approach in microsatellite-stable (MSS) CRC, which is present in 95% of patients. In this review, we focus on metabolic singularities-at the transcriptomic (either bulk or single cell), proteomic, and post-translational modification levels-that induce immunosuppression in cancer and specifically in MSS CRC. First, we evaluate the current efficacy of ICIs in limited and metastatic disease in MSS CRC. Second, we discuss the latest findings on the potential biomarkers for evaluating ICI efficacy in MSS CRC using strict REMARK criteria. Third, we review the current evidence on metabolic patterns in CRC tumors and immune cell metabolism to advance our understanding of metabolic crosstalk and to pave the way for the development of combination strategies to enhance ICI efficacy.
Collapse
Affiliation(s)
- Teresa Gorría
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
- Translational Genomics and Targeted Therapies in Solid Tumors, Agustí Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Medicine Department, University of Barcelona, 08036 Barcelona, Spain
| | - Marina Sierra-Boada
- Medical Oncology Department, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, 08208 Sabadell, Spain;
| | - Mariam Rojas
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
| | - Carolina Figueras
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08036 Barcelona, Spain;
- Institute of Biomedicine of University of Barcelona (IBUB), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Sergio Madurga
- Department of Material Science and Physical Chemistry, Research Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, 08028 Barcelona, Spain;
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08036 Barcelona, Spain;
- Institute of Biomedicine of University of Barcelona (IBUB), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Joan Maurel
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
- Translational Genomics and Targeted Therapies in Solid Tumors, Agustí Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Medicine Department, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
48
|
Corvigno S, Fernebro J, Karlsson JS, Mezheieusky A, Martín-Bernabé A, De La Fuente LM, Westbom-Fremer S, Carlson JW, Klein C, Kannisto P, Hedenfalk I, Malander S, Östman A, Dahlstrand H. High prevalence of FAP+ cancer-associated fibroblasts predicts poor outcome in patients with high-grade serous ovarian cancer with high CD8 T-cell density. Gynecol Oncol 2025; 193:148-155. [PMID: 39914230 DOI: 10.1016/j.ygyno.2025.01.010] [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: 08/13/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 03/03/2025]
Abstract
OBJECTIVE Studies have implied that fibroblasts may act as regulators of immune cells in the tumor microenvironment (TME). We investigated the clinical relevance of fibroblast activation protein (FAP) positive stroma in high-grade serous ovarian cancer (HGSC) in relation to CD8+ lymphocyte's infiltration. METHODS In a discovery cohort (N = 113) of HGSC, expression of FAP and CD8 in the TME was analyzed with immunohistochemistry. Results were correlated with overall survival (OS) and progression-free survival (PFS). The findings were validated in an independent cohort of HGSC (N = 121) and in public available datasets. RESULTS High infiltration of CD8+ cells in the TME of HGSC was found to be associated with longer OS, as previously known. Increased expression of FAP was associated with shorter median PFS (11.4 vs. 18.6 months) in tumors with high density of CD8+ cells (HR 4.03, CI 95 % 1.38-11.72, p = 0.01). Similarly, in the validation cohort, high intensity of FAP in cases with high density of CD8+ cells was associated with shorter OS, 31.5 vs 76.9 months (HR 2.83; 95 % CI 1.17-6.86, p = 0.02). The results were consistent in multivariable analyses. The association between high FAP expression and poor outcome in high density CD8 HGSC was also confirmed in publicly available datasets. CONCLUSIONS The TME infiltration of FAP-positive fibroblasts is associated with poor prognosis in HGSC with high CD8+ cells density. Targeting the FAP+ subset of fibroblasts may unlock the local immune-activation in the TME thus enhance the positive prognostic effect of T-cells in ovarian cancer.
Collapse
Affiliation(s)
- Sara Corvigno
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Josefin Fernebro
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Gynecologic Oncology, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Josefin Severin Karlsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden.
| | - Artur Mezheieusky
- IGP, Uppsala University, Sweden; Vall d'Hebron Institute of Oncology, Molecular oncology group, Barcelona, Spain
| | | | - Laura Martin De La Fuente
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Sofia Westbom-Fremer
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Joseph W Carlson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Pathology and Laboratory Medicine, University of Southern California, Los Angeles, USA
| | | | - Paivi Kannisto
- Department of Obstetrics and Gynecology, Skåne University Hospital and Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and University Hospital, Lund, Sweden
| | - Susanne Malander
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Hanna Dahlstrand
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Gynecologic Oncology, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
49
|
Becherucci G, Ruffolo C, Scarpa M, Scognamiglio F, Stepanyan A, Maretto I, Kotsafti A, De Simoni O, Pilati P, Franzato B, Scapinello A, Bergamo F, Massani M, Stecca T, Pozza A, Cataldo I, Brignola S, Pellegrini V, Fassan M, Guzzardo V, Dal Santo L, Salmaso R, Carlotta C, Dei Tos AP, Angriman I, Spolverato G, Chiminazzo V, Negro S, Vignotto C, Marchegiani F, Facci L, Rivella G, Bao QR, Baldo A, Pucciarelli S, Zizzo M, Businello G, Salmaso B, Parini D, Pirozzolo G, Recordare A, Tagliente G, Bordignon G, Merenda R, Licia L, Pozza G, Godina M, Mondi I, Verdi D, Da Lio C, Guerriero S, Piccioli A, Portale G, Zuin M, Cipollari C, Noaro G, Cola R, Candioli S, Gavagna L, Ricagna F, Ortenzi M, Guerrieri M, Tomassi M, Tedeschi U, Marinelli L, Barbareschi M, Bertalot G, Brolese A, Ceccarini L, Antoniutti M, Porzionato A, Agostini M, Cavallin F, Tussardi G, Di Camillo B, Bardini R, Castagliuolo I, Scarpa M. IMMUNOREACT 8: Immune markers of local tumor spread in patients undergoing transanal excision for clinically N0 rectal cancer. Surgery 2025; 178:108902. [PMID: 39572264 DOI: 10.1016/j.surg.2024.09.043] [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: 06/10/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Transanal excision of rectal cancer can be considered the definitive surgical treatment if the depth spread is T1 or lower, and the lesion is completely included within the resection margin. This study aims to analyze the immune microenvironment in healthy rectal mucosa as a possible predictor of tumor infiltration depth, lateral tumor spread, and recurrence of rectal cancer after transanal local excision. METHODS This study is a subanalysis of data from the IMMUNOREACT 1 and 2 trials (NCT04915326 and NCT04917263, respectively) including all the patients who underwent transanal excision of rectal cancer. This multicentric study collected healthy mucosa surrounding the neoplasms of patients with rectal cancer. A panel of immune markers was investigated at immunohistochemistry: CD3, CD4, CD8, CD8β, Tbet, FoxP3, PD-L1, MSH6, and PMS2 and CD80. Flow cytometry determined the proportion of epithelial cells expressing CD80, CD86, CD40, HLA ABC or HLA DR and the proportion of activated CD8+ T cells, CD4+ Th1 cells, and Treg. RESULTS Receiver operating characteristic curve analysis for predicting deep tumor spread showed an area under the curve of 0.70 (95% confidence interval: 0.60-0.80) for CD25+FoxP3+ cell rate and 0.74 (95% confidence interval: 0.53-0.92) for CK+CD86+ cell rate. Receiver operating characteristic curve analysis for predicting lateral tumor spread showed an area under the curve of 0.82 (95% confidence interval: 0.61-0.99) for CD8+CD38+ MFI, 0.96 (95% confidence interval: 0.85-0.99) for CD8β infiltration, and 0.97 (95% confidence interval: 0.87-0.99) for CK+HLAabc+ cell rate. Receiver operating characteristic curve analysis for predicting recurrence showed an area under the curve of 0.93 (95% confidence interval: 0.76-0.99) for CD8+CD38+ MFI and 0.94 (95% confidence interval: 0.78-0.99) for CD8+CD28+ MFI. Low CD8+CD38+ MFI and low CD8+CD28+ MFI were associated with shorter disease-free survival (P = .025 and P = .021, respectively). CONCLUSION Our study showed that the association between the high proportion of epithelial cells acting as presenting cells and deep or lateral tumor spread may be explained by the presence of a greater tumor load at the site. Moreover, it showed that weak activation of CD8+ T cells within the rectal mucosa is associated with lateral tumor spread and eventually a higher recurrence rate. The mucosal level of CD8β infiltration detected at immunohistochemistry might be tested as a marker of lateral tumor spread and potentially translated into clinical practice.
Collapse
Affiliation(s)
| | - Cesare Ruffolo
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Melania Scarpa
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | | | - Astghik Stepanyan
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Isacco Maretto
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Andromachi Kotsafti
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Ottavia De Simoni
- Oncological Surgery Unit, Veneto Institute of Oncology IOV-IRCCS, Castelfranco Veneto, Italy
| | - Pierluigi Pilati
- Oncological Surgery Unit, Veneto Institute of Oncology IOV-IRCCS, Castelfranco Veneto, Italy
| | - Boris Franzato
- Oncological Surgery Unit, Veneto Institute of Oncology IOV-IRCCS, Castelfranco Veneto, Italy
| | - Antonio Scapinello
- Pathology Unit, Veneto Institute of Oncology IOV-IRCCS, Castelfranco Veneto, Italy
| | - Francesca Bergamo
- Oncology 1 Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Marco Massani
- General Surgery 3 Unit, Azienda ULSS n 2 Marca Trevigiana, Treviso, Italy
| | - Tommaso Stecca
- General Surgery 3 Unit, Azienda ULSS n 2 Marca Trevigiana, Treviso, Italy
| | - Anna Pozza
- General Surgery 3 Unit, Azienda ULSS n 2 Marca Trevigiana, Treviso, Italy
| | - Ivana Cataldo
- Pathology Unit, Azienda ULSS n 2 Marca Trevigiana, Treviso, Italy
| | - Stefano Brignola
- Pathology Unit, Azienda ULSS n 2 Marca Trevigiana, Treviso, Italy
| | | | - Matteo Fassan
- Pathology Unit, Azienda ULSS n 2 Marca Trevigiana, Treviso, Italy
| | - Vincenza Guzzardo
- Pathology Unit, DIMED, Università degli Studi di Padova, Padova, Italy
| | - Luca Dal Santo
- Pathology Unit, DIMED, Università degli Studi di Padova, Padova, Italy
| | - Roberta Salmaso
- Pathology Unit, DIMED, Università degli Studi di Padova, Padova, Italy
| | - Ceccon Carlotta
- Pathology Unit, DIMED, Università degli Studi di Padova, Padova, Italy
| | | | - Imerio Angriman
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Gaya Spolverato
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | | | - Silvia Negro
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Chiara Vignotto
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | | | - Luca Facci
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Giorgio Rivella
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Quoc Riccardo Bao
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Andrea Baldo
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | | | - Maurizio Zizzo
- General Surgery Unit, Arcispedale Santa Maria Nuova IRCCS, Reggio Emilia, Italy
| | | | | | - Dario Parini
- General Surgery Unit, Azienda ULSS n 5 Polesana, Rovigo, Italy
| | | | | | | | | | - Roberto Merenda
- General Surgery Unit, Azienda ULSS 3 Serenissima, Venezia, Italy
| | - Laurino Licia
- Pathology Unit, Azienda ULSS 3 Serenissima, Venezia, Italy
| | - Giulia Pozza
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Mario Godina
- General Surgery Unit, Azienda ULSS 3 Serenissima, Dolo, Italy
| | - Isabella Mondi
- General Surgery Unit, Azienda ULSS 3 Serenissima, MIrano, Italy
| | - Daunia Verdi
- General Surgery Unit, Azienda ULSS 3 Serenissima, MIrano, Italy
| | - Corrado Da Lio
- General Surgery Unit, Azienda ULSS 3 Serenissima, MIrano, Italy
| | | | | | - Giuseppe Portale
- General Surgery Unit, Azienda ULSS 7 Pedemontana, Santorso, Italy
| | - Matteo Zuin
- General Surgery Unit, Azienda ULSS 6 Euganea, Cittadella, Italy
| | | | - Giulia Noaro
- General Surgery Unit, Azienda ULSS 6, Schiavonia, Italy
| | - Roberto Cola
- General Surgery Unit, Azienda ULSS 6, Schiavonia, Italy
| | | | - Laura Gavagna
- General Surgery Unit, Azienda ULSS 1 Dolomiti, Belluno, Italy
| | - Fabio Ricagna
- General Surgery Unit, Azienda ULSS 1 Dolomiti, Belluno, Italy
| | - Monica Ortenzi
- General Surgery Unit, Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona, Italy
| | - Mario Guerrieri
- General Surgery Unit, Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona, Italy
| | | | | | - Laura Marinelli
- General Surgery Unit, Azienda Provinciale per i Servizi Sanitari, Trento, Italy
| | - Mattia Barbareschi
- Pathology Unit, Azienda Provinciale per i Servizi Sanitari, Trento, Italy; Centre for Medical Sciences-CISMed, University of Trento, Italy
| | - Giovanni Bertalot
- Pathology Unit, Azienda Provinciale per i Servizi Sanitari, Trento, Italy; Centre for Medical Sciences-CISMed, University of Trento, Italy
| | - Alberto Brolese
- General Surgery Unit, Azienda Provinciale per i Servizi Sanitari, Trento, Italy
| | | | | | - Andrea Porzionato
- Department of Molecular Medicine, Università degli Studi di Padova, Italy
| | - Marco Agostini
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | | | - Gaia Tussardi
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Barbara Di Camillo
- Department of Information Engineering, Università degli Studi di Padova, Italy
| | - Romeo Bardini
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | | | - Marco Scarpa
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padova, Italy.
| |
Collapse
|
50
|
Müller C, Macher-Beer A, Birnleitner H, Rainer M, Sachet M, Oehler R, Bachleitner-Hofmann T. Effect of systemic FOLFOXIRI plus bevacizumab treatment of colorectal peritoneal metastasis on local and systemic immune cells. Surgery 2025; 178:108868. [PMID: 39472264 DOI: 10.1016/j.surg.2024.09.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/30/2024] [Accepted: 09/18/2024] [Indexed: 01/11/2025]
Abstract
AIM The immune system plays a crucial role in the outcome of colorectal cancer. Systemic chemotherapies modulate the immune cell composition. Little is known about these changes in peritoneal metastasized colorectal cancer. Thus, we aimed to characterize local and systemic immune cells in the course of systemic chemotherapy. METHODS We included in total 20 patients with peritoneal metastasized colorectal cancer in our exploratory study. Initially, we investigated the peripheral blood cell distributions before and after systemic chemotherapy in a set of 11 retrospectively collected samples. Then, a prospective clinical cohort was set up to evaluate local and systemic immune cell distribution in detail (n = 9). Tumor tissue, peritoneal fluid, and peripheral blood were collected. The main immune cell subtypes were characterized using flow cytometry and immunohistochemistry, respectively. RESULTS Neutrophils and the neutrophil-to-lymphocyte ratio significantly declined in response to systemic chemotherapy while circulating T cells increased (CD8+P = .015, CD4+P = .041). In peritoneal fluid, we observed a decrease of CD25+/FOXP3+/CD4+ regulatory T cells (P = .049) without loss of their ability to produce interferon gamma. T-cell infiltration in the tumor microenvironment showed a considerable variability between patients. However, the number of tumor-infiltrating CD8+ lymphocytes was not significantly changed by the application of systemic chemotherapy. Neither tumor cells nor lymphocytes or macrophages showed noteworthy expression of PD1 or PD-L1. CONCLUSION Our data show that immune cell distribution after systemic chemotherapy changes in peripheral blood. Interestingly, in peritoneal fluid only the inhibitory Treg population decreased and local T cells within peritoneal metastases remain unaffected. These data indicate little to no effect of systemic chemotherapy on the local immune system, supporting the need for new therapeutic options.
Collapse
Affiliation(s)
- Catharina Müller
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Austria.
| | | | - Hanna Birnleitner
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Austria
| | - Marlene Rainer
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Austria
| | - Monika Sachet
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Austria
| | - Rudolf Oehler
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Austria
| | | |
Collapse
|