1
|
Zhang L, Liu T, Chen M, Gao S, Staley CA, Yang L, Zhu L. Dual inhibition of oxidative phosphorylation and glycolysis using a hyaluronic acid nanoparticle NOX inhibitor enhanced response to radiotherapy in colorectal cancer. Biomaterials 2025; 323:123437. [PMID: 40449083 DOI: 10.1016/j.biomaterials.2025.123437] [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: 02/13/2025] [Revised: 05/04/2025] [Accepted: 05/25/2025] [Indexed: 06/02/2025]
Abstract
Metabolic reprogramming characterized by mitochondrial dysfunction and increased glycolysis is associated with aggressive tumor biology and poor therapeutic response. The interplays among NADPH oxidase (NOX)-mediated reactive oxygen species, regulation of glycolysis and oxidative phosphorylation (OXPHOS) in cancer cells suggest an opportunity to develop a new cancer therapy. We found that treatment with a hyaluronic acid nanoparticle encapsulated with GKT831 (HANP/GKT831), a NOX1/4 inhibitor, markedly inhibited the proliferation and invasion of cancer cells. Treated tumor cells had reduced levels of mitochondrial ROS, glycolysis, and OXPHOS. The combination of HANP/GKT831 with radiation reduced colony formation and invasion of tumor cells. The combination therapy markedly inhibited the levels of molecules in glycolysis, OXPHOS, and DNA repairing pathways in tumor cells. Systemic administrations of HANP/GKT831 combined with radiotherapy significantly inhibited tumor growth by 84.7 % in a mouse colorectal tumor model. Tumors treated with HANP/GKT831 and radiation had increased DNA damage and apoptotic cell death. Furthermore, the combined therapy increased intratumoral infiltration of activated cytotoxic T cells and M1 macrophages but reduced the levels of immunosuppressive fibroblasts and M2 macrophages. Our results support HANP/GKT831 as a cancer nanotherapeutic agent that induces redox and bioenergy stresses in cancer cells for enhanced therapeutic response to radiotherapy.
Collapse
Affiliation(s)
- Lumeng Zhang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States; Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Tongrui Liu
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States
| | - Minglong Chen
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States; Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Shi Gao
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Charles A Staley
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, United States
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, United States.
| | - Lei Zhu
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, United States; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, United States.
| |
Collapse
|
2
|
Goto N, Agudo J, Yilmaz ÖH. Early immune evasion in colorectal cancer: interplay between stem cells and the tumor microenvironment. Trends Cancer 2025:S2405-8033(25)00112-8. [PMID: 40382216 DOI: 10.1016/j.trecan.2025.04.016] [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: 01/27/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/20/2025]
Abstract
Most colorectal cancers (CRCs) are characterized by a low mutational burden and an immune-cold microenvironment, limiting the efficacy of immune checkpoint blockade (ICB) therapies. While advanced tumors exhibit diverse immune evasion mechanisms, emerging evidence suggests that aspects of immune escape arise much earlier, within precancerous lesions. In this review, we discuss how early driver mutations and epigenetic alterations contribute to the establishment of an immunosuppressive microenvironment in CRC. We also highlight the dynamic crosstalk between cancer cells, stromal niche cells, and immune cells driving immune evasion and liver metastasis. A deeper understanding of these early events may guide the development of more effective preventive and therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Norihiro Goto
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.
| | - Judith Agudo
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Immunology, Harvard Medical School, Boston, MA 02215, USA; Ludwig Center at Harvard, Boston, MA 02215, USA; Parker Institute for Cancer Immunotherapy at Dana-Farber Cancer Institute, Boston, MA 02215, USA; New York Stem Cell Foundation, New York, NY 10019, USA
| | - Ömer H Yilmaz
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Department of Pathology, Beth Israel Deaconess Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
3
|
Zhu B, Chen P, Aminu M, Li JR, Fujimoto J, Tian Y, Hong L, Chen H, Hu X, Li C, Vokes N, Moreira AL, Gibbons DL, Solis Soto LM, Parra Cuentas ER, Shi O, Diao S, Ye J, Rojas FR, Vilar E, Maitra A, Chen K, Navin N, Nilsson M, Huang B, Heeke S, Zhang J, Haymaker CL, Velcheti V, Sterman DH, Kochat V, Padron WI, Alexandrov LB, Wei Z, Le X, Wang L, Fukuoka J, Lee JJ, Wistuba II, Pass HI, Davis M, Hanash S, Cheng C, Dubinett S, Spira A, Rai K, Lippman SM, Futreal PA, Heymach JV, Reuben A, Wu J, Zhang J. Spatial and multiomics analysis of human and mouse lung adenocarcinoma precursors reveals TIM-3 as a putative target for precancer interception. Cancer Cell 2025:S1535-6108(25)00162-X. [PMID: 40345189 DOI: 10.1016/j.ccell.2025.04.003] [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: 09/12/2024] [Revised: 12/31/2024] [Accepted: 04/08/2025] [Indexed: 05/11/2025]
Abstract
How tumor microenvironment shapes lung adenocarcinoma (LUAD) precancer evolution remains poorly understood. Spatial immune profiling of 114 human LUAD and LUAD precursors reveals a progressive increase of adaptive response and a relative decrease of innate immune response as LUAD precursors progress. The immune evasion features align the immune response patterns at various stages. TIM-3-high features are enriched in LUAD precancers, which decrease in later stages. Furthermore, single-cell RNA sequencing (scRNA-seq) and spatial immune and transcriptomics profiling of LUAD and LUAD precursor specimens from 5 mouse models validate high TIM-3 features in LUAD precancers. In vivo TIM-3 blockade at precancer stage, but not at advanced cancer stage, decreases tumor burden. Anti-TIM-3 treatment is associated with enhanced antigen presentation, T cell activation, and increased M1/M2 macrophage ratio. These results highlight the coordination of innate and adaptive immune response/evasion during LUAD precancer evolution and suggest TIM-3 as a potential target for LUAD precancer interception.
Collapse
Affiliation(s)
- Bo Zhu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pingjun Chen
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Muhammad Aminu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jian-Rong Li
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Junya Fujimoto
- Clinical Research Center in Hiroshima, Hiroshima University Hospital, Hiroshima, Japan
| | - Yanhua Tian
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lingzhi Hong
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hong Chen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xin Hu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chenyang Li
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalie Vokes
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andre L Moreira
- Department of Pathology, NYU Langone Health, New York, NY, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa M Solis Soto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin Roger Parra Cuentas
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ou Shi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Songhui Diao
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jie Ye
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Frank R Rojas
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology and Sheikn Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicolas Navin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Monique Nilsson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Beibei Huang
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Simon Heeke
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cara L Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vamsidhar Velcheti
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Daniel H Sterman
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA; Cardiothoracic Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Veena Kochat
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William I Padron
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, USA
| | - Zhubo Wei
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Junya Fukuoka
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | - Mark Davis
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, CA, USA
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Steven Dubinett
- Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Avrum Spira
- Pathology & Laboratory Medicine, and Bioinformatics, Boston University, Boston, MA, USA
| | - Kunal Rai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - P Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jia Wu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4
|
Liu N, Wu T, Han G, Chen M. Exosome-mediated ferroptosis in the tumor microenvironment: from molecular mechanisms to clinical application. Cell Death Discov 2025; 11:221. [PMID: 40328736 PMCID: PMC12056189 DOI: 10.1038/s41420-025-02484-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Ferroptosis in the tumor microenvironment (TME) plays a crucial role in the development, metastasis, immune escape, and drug resistance of various types of cancer. A better understanding of ferroptosis in the TME could illuminate novel aspects of this process and promote the development of targeted therapies. Compelling evidence indicates that exosomes are key mediators in regulating the TME. In this respect, it is now understood that exosomes can deliver biologically functional molecules to recipient cells, influencing cancer progression by reprogramming the metabolism of cancer cells and their surrounding stromal cells through ferroptosis. In this review, we focus on the role of exosomes in the TME and describe how they contribute to tumor reprogramming, immunosuppression, and the formation of pre-metastatic niches through ferroptosis. In addition, we highlight exosome-mediated ferroptosis as a potential target for cancer therapy and discuss strategies employing exosomes in ferroptosis treatment. Finally, we outline the current applications and challenges of targeted exosome-mediated ferroptosis therapy in tumor immunotherapy and chemotherapy. Our aim is to advance research on the link between exosomes and ferroptosis in the TME, and we pose questions to guide future studies in this area.
Collapse
Affiliation(s)
- Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Tianqing Wu
- XJTLU Wisdom Lake Academy of Pharmacy, Suzhou, Jiangsu Province, China
| | - Guohu Han
- Department of Oncology, Jingjiang People's Hospital Affiliated with Yangzhou University, Jingjiang, China
| | - Minbin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
5
|
Admasu TD, Yu JS. Harnessing Immune Rejuvenation: Advances in Overcoming T Cell Senescence and Exhaustion in Cancer Immunotherapy. Aging Cell 2025; 24:e70055. [PMID: 40178455 PMCID: PMC12073907 DOI: 10.1111/acel.70055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/15/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
Immunotherapy has transformed the landscape of cancer treatment, with T cell-based strategies at the forefront of this revolution. However, the durability of these responses is frequently undermined by two intertwined phenomena: T cell exhaustion and senescence. While exhaustion is driven by chronic antigen exposure in the immunosuppressive tumor microenvironment, leading to a reversible state of diminished functionality, senescence reflects a more permanent, age- or stress-induced arrest in cellular proliferation and effector capacity. Together, these processes represent formidable barriers to sustained anti-tumor immunity. In this review, we dissect the molecular underpinnings of T cell exhaustion and senescence, revealing how these dysfunctions synergistically contribute to immune evasion and resistance across a range of solid tumors. We explore cutting-edge therapeutic approaches aimed at rewiring the exhausted and senescent T cell phenotypes. These include advances in immune checkpoint blockade, the engineering of "armored" CAR-T cells, senolytic therapies that selectively eliminate senescent cells, and novel interventions that reinvigorate the immune system's capacity for tumor eradication. By spotlighting emerging strategies that target both exhaustion and senescence, we provide a forward-looking perspective on the potential to harness immune rejuvenation. This comprehensive review outlines the next frontier in cancer immunotherapy: unlocking durable responses by overcoming the immune system's intrinsic aging and exhaustion, ultimately paving the way for transformative therapeutic breakthroughs.
Collapse
Affiliation(s)
| | - John S. Yu
- Department of NeurosurgeryCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
- Kairos PharmaLos AngelesCaliforniaUSA
| |
Collapse
|
6
|
Cui H, Chen M, Zhao M, Li B. A novel cancer-associated fibroblast-related gene signature for predicting diffuse large B cell lymphoma prognosis using weighted gene co-expression network analysis and machine learning. J Int Med Res 2025; 53:3000605251331250. [PMID: 40279206 PMCID: PMC12035177 DOI: 10.1177/03000605251331250] [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: 11/27/2024] [Accepted: 03/11/2025] [Indexed: 04/27/2025] Open
Abstract
ObjectiveOur objective was to investigate a novel cancer-associated fibroblast-related gene signature for predicting clinical outcomes in patients with diffuse large B cell lymphoma.MethodsThe cancer-associated fibroblast-related module genes were identified from Gene Expression Omnibus datasets using weighted gene co-expression network analysis in our retrospective study. Least Absolute Shrinkage and Selection Operator Cox regression was applied to screen a minimal set of genes and construct a prognostic cancer-associated fibroblast-related gene signature for diffuse large B cell lymphoma. Kaplan-Meier plots and receiver operating characteristic curves were used to assess the prognostic performance of the prognostic cancer-associated fibroblast-related genes. A nomogram encompassing the clinical information and prognostic scores of the patients was constructed. Additionally, the relationships of the gene signature with the immune landscape and drug sensitivity were explored.ResultsCapitalizing on machine learning, we developed a prognostic cancer-associated fibroblast-related gene signature risk model, efficiently categorizing patients with diffuse large B cell lymphoma into high- and low-risk groups and exhibiting a more robust capacity for survival prediction. The nomogram showed stronger prognostic ability than the clinical factor-based model or the risk score alone. We also observed significant differences in immune cell profiles and therapeutic responses between the two groups, offering valuable insights for developing personalized treatments for diffuse large B cell lymphoma.ConclusionsWe developed a prognostic cancer-associated fibroblast-related gene-based genetic risk model to predict the prognosis of diffuse large B cell lymphoma, potentially aiding in treatment selection.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Prognosis
- Machine Learning
- Nomograms
- Gene Expression Regulation, Neoplastic
- Retrospective Studies
- Male
- Female
- Gene Regulatory Networks
- Cancer-Associated Fibroblasts/metabolism
- Cancer-Associated Fibroblasts/pathology
- Biomarkers, Tumor/genetics
- Gene Expression Profiling
- Kaplan-Meier Estimate
- Middle Aged
- Transcriptome
- ROC Curve
Collapse
Affiliation(s)
- Hongxia Cui
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Menglu Chen
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Meifang Zhao
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Bingzong Li
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
7
|
Lan H, Yan W, Huang X, Cui J, Hou H. Multi-omics analysis of the dynamic role of STAR+ cells in regulating platinum-based chemotherapy responses and tumor microenvironment in serous ovarian carcinoma. Front Pharmacol 2025; 16:1545762. [PMID: 40098624 PMCID: PMC11911460 DOI: 10.3389/fphar.2025.1545762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Background Serous ovarian carcinoma (SOC) is the most lethal subtype of ovarian cancer, with chemoresistance to platinum-based chemotherapy remaining a major challenge in improving clinical outcomes. The role of the tumor microenvironment (TME), particularly cancer-associated fibroblasts (CAFs), in modulating chemotherapy responses is not yet fully understood. Methods To explore the relationship between CAF subtypes and chemotherapy sensitivity, we employed single-cell RNA sequencing (scRNA-seq), bulk RNA-seq, spatial transcriptomics, immunohistochemistry (IHC), and immunofluorescence (IF). This multi-omics approach enabled the identification, characterization, and functional analysis of CAF subtypes in both chemotherapy-sensitive and chemotherapy-resistant SOC patients. Results We identified steroidogenic acute regulatory protein-positive (STAR+) cells as a novel CAF subtype enriched in chemotherapy-sensitive SOC patients. STAR + cells exhibited unique transcriptional profiles and were functionally enriched in pathways related to P450 drug metabolism, lipid metabolism, and amino acid metabolism, with enhanced pathway activity observed in chemotherapy-sensitive groups. Spatial transcriptomics and IF revealed that STAR + cells were closely localized to tumor cells, suggesting potential cell-cell interactions. Further communication analysis indicated that STAR + cells may suppress WNT signaling in tumor cells, contributing to improved chemotherapy responses. Importantly, STAR expression levels, validated by IHC, were positively correlated with chemotherapy sensitivity and improved patient prognosis. Platinum-based chemotherapy was shown to increase the proportion of STAR + cells, underscoring their dynamic response to treatment. Conclusion Our study identifies STAR + cells as a novel CAF subtype that enhances chemotherapy sensitivity in SOC. By modulating key metabolic pathways and potentially suppressing WNT signaling, STAR + cells could contribute to improved treatment responses. These findings position STAR + cells as a promising biomarker for predicting chemotherapy efficacy in SOC, which warrants further investigation.
Collapse
Affiliation(s)
- Hongwei Lan
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Weihua Yan
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiao Huang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jiali Cui
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Helei Hou
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
8
|
Zhang Z, Tang Y, Luo D, Qiu J, Chen L. Advances in nanotechnology for targeting cancer-associated fibroblasts: A review of multi-strategy drug delivery and preclinical insights. APL Bioeng 2025; 9:011502. [PMID: 40094065 PMCID: PMC11910205 DOI: 10.1063/5.0244706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) play a crucial role in the tumor microenvironment by promoting tumor growth, immune evasion, and metastasis. Recently, drug delivery systems targeting CAFs have emerged as a promising long-term and effective approach to cancer treatment. Advances in nanotechnology, in particular, have led to the development of nanomedicine delivery systems designed specifically to target CAFs, offering new possibilities for precise and personalized cancer therapies. This article reviews recent progress in drug delivery using nanocarriers that target CAFs. Additionally, we explore the potential of combining multiple therapies, such as chemotherapy and immunotherapy, with nanocarriers to enhance efficacy and overcome drug resistance. Although many preclinical studies show promise, the clinical application of nanomedicine still faces considerable challenges, especially in terms of drug penetration and large-scale production. Therefore, this review aims to provide a fresh perspective on CAF-targeted drug delivery systems and highlight potential future research directions and clinical applications.
Collapse
|
9
|
Caretto M, Gadducci A, Pistolesi S, Mattiussi Tome D, Fanelli GN, Naccarato AG, Simoncini T. Deciphering the stromal molecular landscape: the correlation between p16 and α-SMA in epithelial ovarian cancer. J Cancer Res Clin Oncol 2025; 151:79. [PMID: 39937250 PMCID: PMC11821793 DOI: 10.1007/s00432-025-06120-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025]
Abstract
INTRODUCTION The tumor microenvironment (TME) plays an essential role in promoting cancer initiation, progression and metastasis. Cancer-associated fibroblasts (CAFs) are a major constituent of the TME, but universal CAFs' markers have not yet been identified. We selected several new biomarkers [p16 and α-smooth muscle actin (α-SMA)] to investigate the molecular landscape in epithelial ovarian cancer (EOC), given to the unique TME of this malignancy. METHODS In total, 64 patients with a diagnosis of EOC who underwent primary debulking surgery (PDS) at the Department of Gynecology and Obstetrics of the University of Pisa were enrolled between January 2019 and June 2021. The stromal expression of α-SMA and p16 was investigated by using immunohistochemistry, and the correlations between p16 and α-SMA immunoreactivity and BRCA mutational status were analyzed. RESULTS Positive p16 stromal expression was found in 6 out of 38 (15,78%) patients with wild-type BRCA and in only 1 of the 22 (4,50%) patients with mutated BRCA. Conversely, positive α-SMA expression was detected in 34 of 38 patients with wild-type BRCA (89,47%) and in 21 of 22 patients (95,45%) with mutated BRCA. There was a significant difference (r = -0,32) between the negative stromal p16 expression and the positive stromal expression of α-SMA. CONCLUSION This study suggests a new correlation between stromal expression of p16 and α-SMA and BRCA mutational status in EOC. Further investigations are strongly warranted to improve the understanding of the landscape of this malignancy.
Collapse
Affiliation(s)
- Marta Caretto
- Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology I, University of Pisa, Pisa, Italy.
| | - Angiolo Gadducci
- Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology I, University of Pisa, Pisa, Italy
| | - Sabina Pistolesi
- First Division of Pathology, Department of Laboratory Medicine, Pisa University Hospital, Pisa, Italy
| | - Diletta Mattiussi Tome
- Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology I, University of Pisa, Pisa, Italy
| | - Giuseppe Nicolò Fanelli
- First Division of Pathology, Department of Laboratory Medicine, Pisa University Hospital, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Antonio Giuseppe Naccarato
- First Division of Pathology, Department of Laboratory Medicine, Pisa University Hospital, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Tommaso Simoncini
- Department of Clinical and Experimental Medicine, Division of Obstetrics and Gynecology I, University of Pisa, Pisa, Italy
| |
Collapse
|
10
|
Cheng PSW, Zaccaria M, Biffi G. Functional heterogeneity of fibroblasts in primary tumors and metastases. Trends Cancer 2025; 11:135-153. [PMID: 39674792 DOI: 10.1016/j.trecan.2024.11.005] [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: 08/29/2024] [Revised: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 12/16/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are abundant components of the tumor microenvironment (TME) of most solid malignancies and have emerged as key regulators of cancer progression and therapy response. Although recent technological advances have uncovered substantial CAF molecular heterogeneity at the single-cell level, defining functional roles for most described CAF populations remains challenging. With the aim of bridging CAF molecular and functional heterogeneity, this review focuses on recently identified functional interactions of CAF subtypes with malignant cells, immune cells, and other stromal cells in primary tumors and metastases. Dissecting the heterogeneous functional crosstalk of specific CAF populations with other components is starting to uncover candidate combinatorial strategies for therapeutically targeting the TME and cancer progression.
Collapse
Affiliation(s)
- Priscilla S W Cheng
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Marta Zaccaria
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Giulia Biffi
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK.
| |
Collapse
|
11
|
Li X, Gao Z, Yang M, Yang C, Yang D, Cui W, Wu D, Zhou J. Unraveling the metastatic niche in breast cancer bone metastasis through single-cell RNA sequencing. Clin Transl Oncol 2025; 27:671-686. [PMID: 39066875 DOI: 10.1007/s12094-024-03594-2] [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: 02/23/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE Breast cancer (BRCA) is characterized by a unique metastatic pattern, often presenting with bone metastasis (BoM), posing significant clinical challenges. Through the study of the immune microenvironment in BRCA BoM offer perspectives for therapeutic interventions targeting this specific metastatic manifestation of BRCA. METHODS This study employs single-cell RNA sequencing and TCGA data analysis to comprehensively compare primary tumors (PT), lymph node metastasis (LN), and BoM. RESULTS AND CONCLUSIONS Our investigation identifies a metastatic niche in BoM marked by an increased abundance of cancer-associated fibroblasts (CAFs) and reduced immune cell presence. A distinct subtype (State 1) of BRCA BoM cells associated with adverse prognosis is identified. State 1, displaying heightened stemness traits, may represent an initiation phase for BoM in BRCA. Complex cell communications involving tumor, stromal, and immune cells are revealed. Interactions of FN1, SPP1, and MDK correlate with elevated immune cells in BoM. CD46, MDK, and PTN interactions drive myofibroblast activation and proliferation, contributing to tissue remodeling. Additionally, MDK, PTN, and FN1 interactions influence FAP+ CAF activation, impacting cell adhesion and migration in BoM. These insights deepen our understanding of the metastatic niche in breast cancer BoM.
Collapse
Affiliation(s)
- Xiangyu Li
- The Stem Cell and Tissue Engineering Research Center, College of Pharmacy, Changzhi Medical College, Changzhi, 046000, Shanxi, People's Republic of China.
| | - Ziyu Gao
- The Stem Cell and Tissue Engineering Research Center, College of Pharmacy, Changzhi Medical College, Changzhi, 046000, Shanxi, People's Republic of China
| | - Meiling Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Ciqiu Yang
- Medical Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Dongyang Yang
- Medical Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Wenhui Cui
- The Stem Cell and Tissue Engineering Research Center, College of Pharmacy, Changzhi Medical College, Changzhi, 046000, Shanxi, People's Republic of China
| | - Dandan Wu
- Department of Breast Oncology Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Jie Zhou
- Department of Breast Oncology Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, Guangdong, China.
| |
Collapse
|
12
|
Ricci JE. Tumor-induced metabolic immunosuppression: Mechanisms and therapeutic targets. Cell Rep 2025; 44:115206. [PMID: 39798090 DOI: 10.1016/j.celrep.2024.115206] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025] Open
Abstract
Metabolic reprogramming in both immune and cancer cells plays a crucial role in the antitumor immune response. Recent studies indicate that cancer metabolism not only sustains carcinogenesis and survival via altered signaling but also modulates immune cell function. Metabolic crosstalk within the tumor microenvironment results in nutrient competition and acidosis, thereby hindering immune cell functionality. Interestingly, immune cells also undergo metabolic reprogramming that enables their proliferation, differentiation, and effector functions. This review highlights the regulation of antitumor immune responses through metabolic reprogramming in cancer and immune cells and explores therapeutic strategies that target these metabolic pathways in cancer immunotherapy, including using chimeric antigen receptor (CAR)-T cells. We discuss innovative combinations of immunotherapy, cellular therapies, and metabolic interventions that could optimize the efficacy of existing treatment protocols.
Collapse
Affiliation(s)
- Jean-Ehrland Ricci
- Université Côte d'Azur, INSERM, C3M, Nice, France; Équipe labellisée LIGUE Contre le Cancer, Nice, France.
| |
Collapse
|
13
|
Xie Z, Cai D, Ye R, Shan Z, Lin Y, Gao F, Shao N, Kuang X. Integrated immune-related gene signature predicts clinical outcome for patients with Luminal B breast cancer. Gland Surg 2024; 13:2253-2263. [PMID: 39822362 PMCID: PMC11733651 DOI: 10.21037/gs-24-377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/03/2024] [Indexed: 01/19/2025]
Abstract
Background Luminal B breast cancer is routinely treated with chemotherapy and endocrine therapy. However, its sensitivity to treatment remains heterogeneous; therefore, identifying patients who may most benefit remains crucial. Immune-related genes are reportedly related to the prognosis of breast cancer. The purpose of this study was to evaluate the impact of an immune-related gene signature (IRGS) in predicting the prognosis of patients with Luminal B breast cancer. Methods We selected patients with Luminal B breast cancer from two large datasets: 488 from the Metabric dataset (training cohort) and 250 patients from The Cancer Genome Atlas (TCGA) dataset (validation cohort). Prognostic analysis was performed to test the predictive value of IRGS, and enrichment analysis and ESTIMATE were used for deeper function analysis. Results A prognostic IRGS model containing 12 immune-related genes was developed. After which, we separated patients with Luminal B breast cancer into low- and high-risk groups in terms of disease-free survival (DFS) (P<0.001). Multivariate analysis identified IRGS as an independent prognostic factor. Furthermore, functional analysis showed that the 12 genes were mainly enriched in pathways related to chemotherapy response, whose expression levels showed completely opposing trends in low- and high-risk groups. Conclusions The novel IRGS is a satisfactory and reliable biomarker to predict the clinical outcome of patients with Luminal B breast cancer which potentially facilitating individualised management. Further studies are needed to assess the clinical potential in predicting prognosis and the treatment options for Luminal B breast cancer patients.
Collapse
Affiliation(s)
- Zhen Xie
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Du Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Runyi Ye
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen Shan
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ying Lin
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Feng Gao
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Nan Shao
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaying Kuang
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Ankill J, Zhao Z, Tekpli X, Kure EH, Kristensen VN, Mathelier A, Fleischer T. Integrative pan-cancer analysis reveals a common architecture of dysregulated transcriptional networks characterized by loss of enhancer methylation. PLoS Comput Biol 2024; 20:e1012565. [PMID: 39556603 DOI: 10.1371/journal.pcbi.1012565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024] Open
Abstract
Aberrant DNA methylation contributes to gene expression deregulation in cancer. However, these alterations' precise regulatory role and clinical implications are still not fully understood. In this study, we performed expression-methylation Quantitative Trait Loci (emQTL) analysis to identify deregulated cancer-driving transcriptional networks linked to CpG demethylation pan-cancer. By analyzing 33 cancer types from The Cancer Genome Atlas, we identified and confirmed significant correlations between CpG methylation and gene expression (emQTL) in cis and trans, both across and within cancer types. Bipartite network analysis of the emQTL revealed groups of CpGs and genes related to important biological processes involved in carcinogenesis including proliferation, metabolism and hormone-signaling. These bipartite communities were characterized by loss of enhancer methylation in specific transcription factor binding regions (TFBRs) and the CpGs were topologically linked to upregulated genes through chromatin loops. Penalized Cox regression analysis showed a significant prognostic impact of the pan-cancer emQTL in many cancer types. Taken together, our integrative pan-cancer analysis reveals a common architecture where hallmark cancer-driving functions are affected by the loss of enhancer methylation and may be epigenetically regulated.
Collapse
Affiliation(s)
- Jørgen Ankill
- Department of Cancer Genetics, Institute of Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Zhi Zhao
- Department of Cancer Genetics, Institute of Cancer Research, Oslo University Hospital, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Xavier Tekpli
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Elin H Kure
- Department of Cancer Genetics, Institute of Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Vessela N Kristensen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Anthony Mathelier
- Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Norway, Oslo, Norway
| | - Thomas Fleischer
- Department of Cancer Genetics, Institute of Cancer Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
15
|
郗 雪, 邓 婷, 杜 伯. [Colorectal fibroblasts promote malignant phenotype of colorectal cancer cells by activating the ERK signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1866-1873. [PMID: 39523086 PMCID: PMC11526459 DOI: 10.12122/j.issn.1673-4254.2024.10.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE To investigate the effect of human colorectal fibroblast (CCD-18Co)-conditioned medium (CCD18-Co-CM) on biological behaviors of colorectal cancer (CRC) cells and explore the possible molecular mechanisms. METHODS Real-time cellular analysis (RTCA), clone formation assay and wound healing assay were used to analyze the changes in proliferation, clone formation, and migration abilities of CRC cell lines HCT116 and Caco-2 treated with CCD18-Co-CM. Western blotting was used to detect the changes in ATK, ERK and STAT3 signaling pathways in the CRC cells activated by CCD18-Co-CM. The effect of CCD18-Co-CM on spheroidization ability of the cells was assessed with sphere-formation assay, and the changes in expressions of CRC stemness markers were detected using RT-PCR. RESULTS CCD-18Co-CM significantly promoted proliferation, colony formation, and migration of HCT116 and Caco-2 cells, enhanced sphere-forming ability and expressions of CRC stemness markers, and increased ERK phosphorylation in the cells. Treatment with SCH772984 effectively inhibited CCD-18Co-CM-induced ERK signaling pathway activation, suppressed the malignant phenotype, and lowered the sphere-forming ability and expression of stemness markers of the two CRC cells. CONCLUSION Colorectal fibroblasts promote malignant phenotype of CRC cells by activating the ERK signaling pathway.
Collapse
|
16
|
Wu B, Zhang B, Li B, Wu H, Jiang M. Cold and hot tumors: from molecular mechanisms to targeted therapy. Signal Transduct Target Ther 2024; 9:274. [PMID: 39420203 PMCID: PMC11491057 DOI: 10.1038/s41392-024-01979-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/20/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment, particularly through immune checkpoint blockade (ICB), which has shown notable clinical benefits across various tumor types. Despite the transformative impact of ICB treatment in cancer therapy, only a minority of patients exhibit a positive response to it. In patients with solid tumors, those who respond well to ICB treatment typically demonstrate an active immune profile referred to as the "hot" (immune-inflamed) phenotype. On the other hand, non-responsive patients may exhibit a distinct "cold" (immune-desert) phenotype, differing from the features of "hot" tumors. Additionally, there is a more nuanced "excluded" immune phenotype, positioned between the "cold" and "hot" categories, known as the immune "excluded" type. Effective differentiation between "cold" and "hot" tumors, and understanding tumor intrinsic factors, immune characteristics, TME, and external factors are critical for predicting tumor response and treatment results. It is widely accepted that ICB therapy exerts a more profound effect on "hot" tumors, with limited efficacy against "cold" or "altered" tumors, necessitating combinations with other therapeutic modalities to enhance immune cell infiltration into tumor tissue and convert "cold" or "altered" tumors into "hot" ones. Therefore, aligning with the traits of "cold" and "hot" tumors, this review systematically delineates the respective immune characteristics, influencing factors, and extensively discusses varied treatment approaches and drug targets based on "cold" and "hot" tumors to assess clinical efficacy.
Collapse
Affiliation(s)
- Bo Wu
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bo Zhang
- Department of Youth League Committee, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bowen Li
- Department of Pancreatic and Gastrointestinal Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Haoqi Wu
- Department of Gynaecology and Obstetrics, The Second Hospital of Dalian Medical University, Dalian, China
| | - Meixi Jiang
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
17
|
Liu Y, Liang J, Zhang Y, Guo Q. Drug resistance and tumor immune microenvironment: An overview of current understandings (Review). Int J Oncol 2024; 65:96. [PMID: 39219258 PMCID: PMC11387120 DOI: 10.3892/ijo.2024.5684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
The use of antitumor drugs represents a reliable strategy for cancer therapy. Unfortunately, drug resistance has become increasingly common and contributes to tumor metastasis and local recurrence. The tumor immune microenvironment (TME) consists of immune cells, cytokines and immunomodulators, and collectively they influence the response to treatment. Epigenetic changes including DNA methylation and histone modification, as well as increased drug exportation have been reported to contribute to the development of drug resistance in cancers. In the past few years, the majority of studies on tumors have only focused on the development and progression of a tumor from a mechanistic standpoint; few studies have examined whether the changes in the TME can also affect tumor growth and drug resistance. Recently, emerging evidence have raised more concerns regarding the role of TME in the development of drug resistance. In the present review, it was discussed how the suppressive TME adapts to drug resistance characterized by the cooperation of immune cells, cytokines, immunomodulators, stromal cells and extracellular matrix. Furthermore, it was reviewed how these immunological or metabolic changes alter immuno‑surveillance and thus facilitate tumor drug resistance. In addition, potential targets present in the TME for developing novel therapeutic strategies to improve individualized therapy for cancer treatment were revealed.
Collapse
Affiliation(s)
- Yan Liu
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Jun Liang
- Department of Radiology, Qingdao Haici Hospital, Qingdao, Shandong 266000, P.R. China
| | - Yanping Zhang
- Department of Radiology, Qingdao Haici Hospital, Qingdao, Shandong 266000, P.R. China
| | - Qie Guo
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
18
|
Ye YH, Xin HY, Li JL, Li N, Pan SY, Chen L, Pan JY, Hu ZQ, Wang PC, Luo CB, Sun RQ, Fan J, Zhou J, Zhou ZJ, Zhou SL. Development and validation of a stromal-immune signature to predict prognosis in intrahepatic cholangiocarcinoma. Clin Mol Hepatol 2024; 30:914-928. [PMID: 39103994 PMCID: PMC11540385 DOI: 10.3350/cmh.2024.0296] [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: 04/25/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUNDS/AIMS Intrahepatic cholangiocarcinoma (ICC) is a highly desmoplastic tumor with poor prognosis even after curative resection. We investigated the associations between the composition of the ICC stroma and immune cell infiltration and aimed to develop a stromal-immune signature to predict prognosis in surgically treated ICC. METHODS We recruited 359 ICC patients and performed immunohistochemistry to detect α-smooth muscle actin (α-SMA), CD3, CD4, CD8, Foxp3, CD68, and CD66b. Aniline was used to stain collagen deposition. Survival analyses were performed to detect prognostic values of these markers. Recursive partitioning for a discrete-time survival tree was applied to define a stromal-immune signature with distinct prognostic value. We delineated an integrated stromal-immune signature based on immune cell subpopulations and stromal composition to distinguish subgroups with different recurrence-free survival (RFS) and overall survival (OS) time. RESULTS We defined four major patterns of ICC stroma composition according to the distributions of α-SMA and collagen: dormant (α-SMAlow/collagenhigh), fibrogenic (α-SMAhigh/collagenhigh), inert (α-SMAlow/collagenlow), and fibrolytic (α-SMAhigh/collagenlow). The stroma types were characterized by distinct patterns of infiltration by immune cells. We divided patients into six classes. Class I, characterized by high CD8 expression and dormant stroma, displayed the longest RFS and OS, whereas Class VI, characterized by low CD8 expression and high CD66b expression, displayed the shortest RFS and OS. The integrated stromal-immune signature was consolidated in a validation cohort. CONCLUSION We developed and validated a stromal-immune signature to predict prognosis in surgically treated ICC. These findings provide new insights into the stromal-immune response to ICC.
Collapse
Affiliation(s)
- Yu-Hang Ye
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao-Yang Xin
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia-Li Li
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ning Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Si-Yuan Pan
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Long Chen
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing-Yue Pan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhi-Qiang Hu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng-Cheng Wang
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chu-Bin Luo
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rong-Qi Sun
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng-Jun Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shao-Lai Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Takei J, Maeda M, Fukasawa N, Kawashima M, Miyake M, Tomoto K, Nawate S, Teshigawara A, Suzuki T, Yamamoto Y, Nagashima H, Mori R, Fukushima R, Matsushima S, Kino H, Muroi A, Tsurubuchi T, Sakamoto N, Nishiwaki K, Yano S, Hasegawa Y, Murayama Y, Akasaki Y, Shimoda M, Ishikawa E, Tanaka T. Comparative analyses of immune cells and alpha-smooth muscle actin-positive cells under the immunological microenvironment between with and without dense fibrosis in primary central nervous system lymphoma. Brain Tumor Pathol 2024; 41:97-108. [PMID: 39186169 PMCID: PMC11499374 DOI: 10.1007/s10014-024-00488-7] [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: 02/24/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Histopathologic examinations of primary central nervous system lymphoma (PCNSL) reveal concentric accumulation of lymphocytes in the perivascular area with fibrosis. However, the nature of this fibrosis in "stiff" PCNSL remains unclear. We have encountered some PCNSLs with hard masses as surgical findings. This study investigated the dense fibrous status and tumor microenvironment of PCNSLs with or without stiffness. We evaluated by silver-impregnation nine PCNSLs with stiffness and 26 PCNSLs without stiffness. Six of the nine stiff PCNSLs showed pathological features of prominent fibrosis characterized by aggregation of reticulin fibers, and collagen accumulations. Alpha-smooth muscle actin (αSMA)-positive spindle cells as a cancer-associated fibroblast, the populations of T lymphocytes, and macrophages were compared between fibrous and control PCNSLs. Fibrous PCNSLs included abundant αSMA-positive cells in both intra- and extra-tumor environments (5/6, 87% and 3/6, 50%, respectively). Conversely, only one out of the seven control PCNSL contained αSMA-positive cells in the intra-tumoral area. Furthermore, the presence of extra-tumoral αSMA-positive cells was associated with infiltration of T lymphocytes and macrophages. In conclusion, recognizing the presence of dense fibrosis in PCNSL can provide insights into the tumor microenvironment. These results may help stratify patients with PCNSL and improve immunotherapies for these patients.
Collapse
Affiliation(s)
- Jun Takei
- Department of Neurosurgery, The Jikei University Katsushika Medical Center, 6-41-2 Aoto, Katsushika-ku, Tokyo, 125-8506, Japan
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Miku Maeda
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Nei Fukasawa
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Masaharu Kawashima
- Division of Clinical Oncology and Hematology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Misayo Miyake
- Department of Pathology, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Kyoichi Tomoto
- Department of Neurosurgery, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Shohei Nawate
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Akihiko Teshigawara
- Department of Neurosurgery, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Tomoya Suzuki
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Yohei Yamamoto
- Department of Neurosurgery, The Jikei University Daisan Hospital, 4-11-1 Izumi-honcho, Komae-shi, Tokyo, 201-8601, Japan
| | - Hiroyasu Nagashima
- Department of Neurosurgery, The Jikei University Katsushika Medical Center, 6-41-2 Aoto, Katsushika-ku, Tokyo, 125-8506, Japan
| | - Ryosuke Mori
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Ryoko Fukushima
- Division of Clinical Oncology and Hematology, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Satoshi Matsushima
- Department of Radiology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hiroyoshi Kino
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Ai Muroi
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Takao Tsurubuchi
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Noriaki Sakamoto
- Department of Clinical Pathology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kaichi Nishiwaki
- Division of Clinical Oncology and Hematology, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yuzuru Hasegawa
- Department of Neurosurgery, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Yuichi Murayama
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Yasuharu Akasaki
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Masayuki Shimoda
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Eiichi Ishikawa
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Toshihide Tanaka
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan.
- Department of Neurosurgery, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan.
| |
Collapse
|
20
|
Schneider P, Zhang H, Simic L, Dai Z, Schrörs B, Akilli-Öztürk Ö, Lin J, Durak F, Schunke J, Bolduan V, Bogaert B, Schwiertz D, Schäfer G, Bros M, Grabbe S, Schattenberg JM, Raemdonck K, Koynov K, Diken M, Kaps L, Barz M. Multicompartment Polyion Complex Micelles Based on Triblock Polypept(o)ides Mediate Efficient siRNA Delivery to Cancer-Associated Fibroblasts for Antistromal Therapy of Hepatocellular Carcinoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404784. [PMID: 38958110 DOI: 10.1002/adma.202404784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/27/2024] [Indexed: 07/04/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer and the third leading cause for cancer-related death worldwide. The tumor is difficult-to-treat due to its inherent resistance to chemotherapy. Antistromal therapy is a novel therapeutic approach, targeting cancer-associated fibroblasts (CAF) in the tumor microenvironment. CAF-derived microfibrillar-associated protein 5 (MFAP-5) is identified as a novel target for antistromal therapy of HCC with high translational relevance. Biocompatible polypept(o)ide-based polyion complex micelles (PICMs) constructed with a triblock copolymer composed of a cationic poly(l-lysine) complexing anti-MFAP-5 siRNA (siMFAP-5) via electrostatic interaction, a poly(γ-benzyl-l-glutamate) block loading cationic amphiphilic drug desloratatine (DES) via π-π interaction as endosomal escape enhancer and polysarcosine poly(N-methylglycine) for introducing stealth properties, are generated for siRNA delivery. Intravenous injection of siMFAP-5/DES PICMs significantly reduces the hepatic tumor burden in a syngeneic implantation model of HCC, with a superior MFAP-5 knockdown effect over siMFAP-5 PICMs or lipid nanoparticles. Transcriptome and histological analysis reveal that MFAP-5 knockdown inhibited CAF-related tumor vascularization, suggesting the anti-angiogenic effect of RNA interference therapy. In conclusion, multicompartment PICMs combining siMFAP-5 and DES in a single polypept(o)ide micelle induce a specific knockdown of MFAP-5 and demonstrate a potent antitumor efficacy (80% reduced tumor burden vs untreated control) in a clinically relevant HCC model.
Collapse
Affiliation(s)
- Paul Schneider
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128, Mainz, Germany
| | - Heyang Zhang
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2333CC, Netherlands
| | - Leon Simic
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128, Mainz, Germany
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2333CC, Netherlands
| | - Zhuqing Dai
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2333CC, Netherlands
| | - Barbara Schrörs
- Biosampling Unit, TRON gGmbH - Translational Oncology at the University Medical Center of the Johannes Gutenberg University, Freiligrathstr. 12, 55131, Mainz, Germany
| | - Özlem Akilli-Öztürk
- Biosampling Unit, TRON gGmbH - Translational Oncology at the University Medical Center of the Johannes Gutenberg University, Freiligrathstr. 12, 55131, Mainz, Germany
| | - Jian Lin
- Max Planck Institute for Polymer Research, Physics at Interphases, Ackermannweg 10, 55128, Mainz, Germany
| | - Feyza Durak
- Biosampling Unit, TRON gGmbH - Translational Oncology at the University Medical Center of the Johannes Gutenberg University, Freiligrathstr. 12, 55131, Mainz, Germany
| | - Jenny Schunke
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128, Mainz, Germany
| | - Vanessa Bolduan
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128, Mainz, Germany
| | - Bram Bogaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - David Schwiertz
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128, Mainz, Germany
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2333CC, Netherlands
| | - Gabriela Schäfer
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128, Mainz, Germany
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2333CC, Netherlands
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128, Mainz, Germany
| | - Jörn Markus Schattenberg
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421, Homburg, Germany
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research, Physics at Interphases, Ackermannweg 10, 55128, Mainz, Germany
| | - Mustafa Diken
- Biosampling Unit, TRON gGmbH - Translational Oncology at the University Medical Center of the Johannes Gutenberg University, Freiligrathstr. 12, 55131, Mainz, Germany
| | - Leonard Kaps
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128, Mainz, Germany
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421, Homburg, Germany
| | - Matthias Barz
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128, Mainz, Germany
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2333CC, Netherlands
| |
Collapse
|
21
|
Zhu Z, Li J, Fa Z, Xu X, Wang Y, Zhou J, Xu Y. Functional gene signature offers a powerful tool for characterizing clinicopathological features and depicting tumor immune microenvironment of colorectal cancer. BMC Cancer 2024; 24:1199. [PMID: 39342165 PMCID: PMC11437988 DOI: 10.1186/s12885-024-12996-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Colorectal cancer, a prevalent malignancy worldwide, poses a significant challenge due to the lack of effective prognostic tools. In this study, we aimed to develop a functional gene signature to stratify colorectal cancer patients into different groups with distinct characteristics, which will greatly facilitate disease prediction. RESULTS Patients were stratified into high- and low-risk groups using a prediction model built based on the functional gene signature. This innovative approach not only predicts clinicopathological features but also reveals tumor immune microenvironment types and responses to immunotherapy. The study reveals that patients in the high-risk group exhibit poorer pathological features, including invasion depth, lymph node metastasis, and distant metastasis, as well as unfavorable survival outcomes in terms of overall survival and disease-free survival. The underlying mechanisms for these observations are attributed to upregulated tumor-related signaling pathways, increased infiltration of pro-tumor immune cells, decreased infiltration of anti-tumor immune cells, and a lower tumor mutation burden. Consequently, patients in the high-risk group exhibit a diminished response to immunotherapy. Furthermore, the high-risk group demonstrates enrichment in extracellular matrix-related functions and significant infiltration of cancer-associated fibroblasts (CAFs). Single-cell transcriptional data analysis identifies CAFs as the primary cellular type expressing hub genes, namely ACTA2, TPM2, MYL9, and TAGLN. This finding is further validated through multiple approaches, including multiplex immunohistochemistry (mIHC), polymerase chain reaction (PCR), and western blot analysis. Notably, TPM2 emerges as a potential biomarker for identifying CAFs in colorectal cancer, distinguishing them from both colorectal cancer cell lines and normal colon epithelial cell lines. Co-culture of CAFs and colorectal cancer cells revealed that CAFs could enhance the tumorigenic biofunctions of cancer cells indirectly, which could be partially inhibited by knocking down CAF original TPM2 expression. CONCLUSIONS This study introduces a functional gene signature that effectively and reliably predicts clinicopathological features and the tumor immune microenvironment in colorectal cancer. Moreover, the identification of TPM2 as a potential biomarker for CAFs holds promising implications for future research and clinical applications in the field of colorectal cancer.
Collapse
Affiliation(s)
- Ziyan Zhu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jikun Li
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenzhong Fa
- Department of General Surgery, Wujin Hospital Affiliated With Jiangsu University, Changzhou, Jiangsu Province, China
- Department of General Surgery, the Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu Province, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou, Jiangsu Province, China
| | - Xuezhong Xu
- Department of General Surgery, Wujin Hospital Affiliated With Jiangsu University, Changzhou, Jiangsu Province, China
- Department of General Surgery, the Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu Province, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou, Jiangsu Province, China
| | - Yue Wang
- Department of General Surgery, Wujin Hospital Affiliated With Jiangsu University, Changzhou, Jiangsu Province, China
- Department of General Surgery, the Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu Province, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou, Jiangsu Province, China
| | - Jie Zhou
- Department of General Surgery, Wujin Hospital Affiliated With Jiangsu University, Changzhou, Jiangsu Province, China
- Department of General Surgery, the Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu Province, China
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou, Jiangsu Province, China
| | - Yixin Xu
- Department of General Surgery, Wujin Hospital Affiliated With Jiangsu University, Changzhou, Jiangsu Province, China.
- Department of General Surgery, the Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu Province, China.
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou, Jiangsu Province, China.
| |
Collapse
|
22
|
Shao M, Gao Y, Xu X, Chan DW, Du J. Exosomes: Key Factors in Ovarian Cancer Peritoneal Metastasis and Drug Resistance. Biomolecules 2024; 14:1099. [PMID: 39334866 PMCID: PMC11430201 DOI: 10.3390/biom14091099] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Ovarian cancer remains a leading cause of death among gynecological cancers, largely due to its propensity for peritoneal metastasis and the development of drug resistance. This review concentrates on the molecular underpinnings of these two critical challenges. We delve into the role of exosomes, the nano-sized vesicles integral to cellular communication, in orchestrating the complex interactions within the tumor microenvironment that facilitate metastatic spread and thwart therapeutic efforts. Specifically, we explore how exosomes drive peritoneal metastasis by promoting epithelial-mesenchymal transition in peritoneal mesothelial cells, altering the extracellular matrix, and supporting angiogenesis, which collectively enable the dissemination of cancer cells across the peritoneal cavity. Furthermore, we dissect the mechanisms by which exosomes contribute to the emergence of drug resistance, including the sequestration and expulsion of chemotherapeutic agents, the horizontal transfer of drug resistance genes, and the modulation of critical DNA repair and apoptotic pathways. By shedding light on these exosome-mediated processes, we underscore the potential of exosomal pathways as novel therapeutic targets, offering hope for more effective interventions against ovarian cancer's relentless progression.
Collapse
Affiliation(s)
- Ming Shao
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
- Department of Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen 518172, China
| | - Yunran Gao
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Xiling Xu
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - David Wai Chan
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Juan Du
- Department of Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen 518172, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
23
|
Weng D, Guo R, Dong C, Luo Y, Qiu D, Xu L, Xu G. Magnetic Resonance Imaging of Fibroblast Activation Protein Using a Targeted Gadolinium-Based Contrast Agent. Mol Pharm 2024. [PMID: 39159402 DOI: 10.1021/acs.molpharmaceut.3c01250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
The aim of this study was to synthesize a quinoline-based MRI contrast agent, Gd-DOTA-FAPI04, and assess its capacity for targeting fibroblast activation protein (FAP)-positive tumors in vivo. Gd-DOTA-FAPI04 was synthesized by attaching a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) complex of gadolinium(III) to FAP inhibitor FAPI04. The longitudinal relaxation time (T1) of the contrast agent was measured using a Siemens Prisma 3.0T MR system, and the CCK-8 assay was performed to evaluate its potential cytotoxicity. Male nude mice bearing tumors grown from FAP-expressing fibrosarcoma cells were divided into experimental (n = 4) and control (n = 4) groups, and T1-weighted image enhancement was measured at different times (0, 10, 30, 60, 90, and 120 min) postinjection of Gd-DOTA-FAPI04. The control group received an additional preinjection of excess FAPI04. FAP expression in tumor tissue was investigated by using immunohistochemistry with an anti-FAP antibody. The longitudinal relaxivities of gadodiamide and Gd-DOTA-FAPI04 were measured to be 3.734 mM-1 s-1 and 5.323 mM-1 s-1, respectively. The CCK-8 assay demonstrated that Gd-DOTA-FAPI04 has minimal toxicity to cultured human fibrosarcoma cells. In vivo MRI showed that peak accumulation of Gd-DOTA-FAPI04 in FAP-expressing tumors occurred 1 h postinjection and could be blocked by preinjection of excess FAPI04. Immunohistochemical analysis of harvested tumor tissue supported the above findings. Gd-DOTA-FAPI04 is a promising contrast agent for in vivo imaging of FAP.
Collapse
Affiliation(s)
- Dinghu Weng
- Department of Medical Imaging, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuchang District, Wuhan 430071, Hubei, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan 430071, Hubei, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, Wuhan 430071, Hubei, China
| | - Rong Guo
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430000, Hubei, China
| | - Changling Dong
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuchang District, Wuhan 430071, Hubei, China
| | - Yuan Luo
- Department of Medical Imaging, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuchang District, Wuhan 430071, Hubei, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan 430071, Hubei, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, Wuhan 430071, Hubei, China
| | - Dasheng Qiu
- Department of Nuclear Medicine, Hubei Cancer Hospital, Wuhan 430079, Hubei, China
| | - Liying Xu
- Department of Medical Imaging, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuchang District, Wuhan 430071, Hubei, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan 430071, Hubei, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, Wuhan 430071, Hubei, China
| | - Guobin Xu
- Department of Medical Imaging, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuchang District, Wuhan 430071, Hubei, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan 430071, Hubei, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, Wuhan 430071, Hubei, China
| |
Collapse
|
24
|
Lapuente-Santana Ó, Sturm G, Kant J, Ausserhofer M, Zackl C, Zopoglou M, McGranahan N, Rieder D, Trajanoski Z, da Cunha Carvalho de Miranda NF, Eduati F, Finotello F. Multimodal analysis unveils tumor microenvironment heterogeneity linked to immune activity and evasion. iScience 2024; 27:110529. [PMID: 39161957 PMCID: PMC11331718 DOI: 10.1016/j.isci.2024.110529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/03/2024] [Accepted: 07/13/2024] [Indexed: 08/21/2024] Open
Abstract
The cellular and molecular heterogeneity of tumors is a major obstacle to cancer immunotherapy. Here, we use a systems biology approach to derive a signature of the main sources of heterogeneity in the tumor microenvironment (TME) from lung cancer transcriptomics. We demonstrate that this signature, which we called iHet, is conserved in different cancers and associated with antitumor immunity. Through analysis of single-cell and spatial transcriptomics data, we trace back the cellular origin of the variability explaining the iHet signature. Finally, we demonstrate that iHet has predictive value for cancer immunotherapy, which can be further improved by disentangling three major determinants of anticancer immune responses: activity of immune cells, immune infiltration or exclusion, and cancer-cell foreignness. This work shows how transcriptomics data can be integrated to derive a holistic representation of the phenotypic heterogeneity of the TME and to predict its unfolding and fate during immunotherapy with immune checkpoint blockers.
Collapse
Affiliation(s)
- Óscar Lapuente-Santana
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Gregor Sturm
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Boehringer Ingelheim International Pharma GmbH & Co KG, 55216 Ingelheim am Rhein, Germany
| | - Joan Kant
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Markus Ausserhofer
- Department of Molecular Biology, Digital Science Center (DiSC), University of Innsbruck, 6020 Innsbruck, Austria
| | - Constantin Zackl
- Department of Molecular Biology, Digital Science Center (DiSC), University of Innsbruck, 6020 Innsbruck, Austria
| | - Maria Zopoglou
- Department of Molecular Biology, Digital Science Center (DiSC), University of Innsbruck, 6020 Innsbruck, Austria
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London WC1E 6DD, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, London WC1E 6DD, UK
| | - Dietmar Rieder
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Zlatko Trajanoski
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | - Federica Eduati
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands
| | - Francesca Finotello
- Department of Molecular Biology, Digital Science Center (DiSC), University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
25
|
Li J, Lin N, Zhang S, Weng L, Chen C, Ou W, Cao Y. Characterization of the tumor microenvironment in breast cancer brain metastasis. Heliyon 2024; 10:e34876. [PMID: 39157383 PMCID: PMC11328047 DOI: 10.1016/j.heliyon.2024.e34876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 08/20/2024] Open
Abstract
Objective The difference in the tumor microenvironment (TME) between primary breast cancer (PBC) and breast cancer brain metastasis (BCBM) is still unknown. Herein, we present the landscape of the TME in PBC and BCBM to better understand the process of BCBM. Methods The Gene Expression Omnibus (GEO) database was used to obtain suitable PBC and BCBM data. Hub genes that were differentially expressed between the two groups were searched. Gene Ontology (GO) and KEGG were used to define the gene's function. Single-cell data were also analyzed to determine the difference between PBC and BCBM. Results Two datasets (GSE100534 and GSE125989) were used to search for hub genes, and 79 genes were either upregulated or downregulated between the two groups. Four hub genes (COL1A1, PDGFR, MMP3 and FZD7) were related to prognosis. GO and KEGG analyses showed that extracellular matrix and focal adhesion play major roles in the metastasis process. Another two datasets (GSE176078 and GSE186344) were enrolled for single-cell analysis. Single-cell analysis demonstrated that immune cells (66.6 %) form the main part of PBC, while cancer-associated fibroblasts (CAFs) (21.7 %) are the main component of BCBM. Immune cell proportion analysis showed that CD4+/CD8+ T cells (28.9 % and 14.3 %, respectively) and macrophages(M2) accounted for the majority of cells in PBC and BCBM, respectively. Further analysis of the classification of CAFs showed that apCAFs were significantly higher in PBC. Conclusions This study presents the landscape of BCBM with hub gene searching and single-cell analysis. Showing the difference in the tumor/immune microenvironment of PBC and BCBM, would be beneficial to explore immunotherapy and targeted therapy for BCBM.
Collapse
Affiliation(s)
- Jingrong Li
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
| | - Nanping Lin
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350001, China
| | - Shengcen Zhang
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
| | - Lihong Weng
- Fujian Cancer Hospital, Fuzhou, Fujian, 350001, China
| | - Chen Chen
- Department of Radiotherapy, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
| | - Wenshi Ou
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350001, China
| | - Yingping Cao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
| |
Collapse
|
26
|
Zhang D, Liang P, Xia B, Wu J, Hu X. Comprehensive pan-cancer analysis of ZNF337 as a potential diagnostic, immunological, and prognostic biomarker. BMC Cancer 2024; 24:987. [PMID: 39123194 PMCID: PMC11313096 DOI: 10.1186/s12885-024-12703-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Zinc Finger Protein 337 (ZNF337) is a novel Zinc Finger (ZNF) protein family member. However, the roles of ZNF337 in human cancers have not yet been investigated. METHODS In this study, with the aid of TCGA databases, GTEx databases, and online websites, we determined the expression levels of ZNF337 in pan-cancer and its potential value as a diagnostic and prognostic marker for pan-cancer and analyzed the relationship between ZNF337 expression and immune cell infiltration and immune checkpoint genes. We then focused our research on the potential of ZNF337 as a biomarker for diagnostic and prognostic in KIRC (kidney renal clear cell carcinoma) and validated in the E-MTAB-1980 database. Moreover, the expression of ZNF337 was detected through qRT-PCR and Western blotting (WB). CCK-8 experiment, colony formation experiment, and EDU experiment were performed to evaluate cell proliferation ability. Wound healing assay and transwell assay were used to analyze its migration ability. The qRT-PCR and WB were used to detect the expression of ZNF337 in tumor tissues and paracancerous tissues of KIRC patients. RESULTS The pan-cancer analysis revealed that abnormal ZNF337 expression was found in multiple human cancer types. ZNF337 had a high diagnostic value in pan-cancer and a significant association with the prognosis of certain cancers, indicating that ZNF337 may be a valuable prognostic biomarker for multiple cancers. Further analysis demonstrated that the expression level of ZNF337 displayed significant correlations with cancer-associated fibroblasts, immune cell infiltration, and immune checkpoint genes in many tumors. Additionally, ZNF337 was observed to have a high expression in KIRC. Its expression was significantly associated with poor prognosis [overall survival (OS), disease-specific survival (DSS)], age, TNM stage, histologic grade, and pathologic stage. The high ZNF337 expression was associated with poor prognosis in the E-MTAB-1980 validation cohort. The in vitro experiments suggested that the expression of ZNF337 in KIRC tumor tissues was higher than in adjacent tissues, and ZNF337 knockdown inhibited the proliferation and migration of KIRC cells, whereas overexpression of ZNF337 had the opposite effects. CONCLUSIONS ZNF337 might be an important prognostic and immunotherapeutic biomarker for pan-cancer, especially in KIRC.
Collapse
Affiliation(s)
- Dongxu Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8 Gongti South Road, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Pu Liang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, 100015, China
| | - Bowen Xia
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8 Gongti South Road, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jitao Wu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Xiaopeng Hu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8 Gongti South Road, Beijing, China.
- Institute of Urology, Capital Medical University, Beijing, China.
| |
Collapse
|
27
|
Meijuan C, Fang M, Qian W. Dachsous cadherin related 1 (DCHS1) is a novel biomarker for immune infiltration and epithelial-mesenchymal transition in endometrial cancer via pan-cancer analysis. J Ovarian Res 2024; 17:162. [PMID: 39123216 PMCID: PMC11312386 DOI: 10.1186/s13048-024-01478-1] [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: 08/11/2023] [Accepted: 07/15/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Dachsous cadherin related 1 (DCHS1) is one of calcium-dependent adhesion membrane proteins and is mainly involved in the development of mammalian tissues. There is a lack of more detailed research on the biological function of DCHS1 in pan-cancer. MATERIALS AND METHODS We evaluated the expression, the prognostic value, the diagnostic value and genomic alterations of DCHS1 by using the databases, including TCGA, UALCAN, HPA, GEPIA2.0 and GSCA. We employed the databases of UCSC, TIMER2.0, TISIDB, GSCA to analyze the association between DCHS1 expression and the immune microenvironment, stemness, TMB, MSI and anticancer drug sensitivity. BioGRID, STRING and GEPIA2.0 were used to perform protein interaction and functional enrichment analysis. Real-time quantitative PCR, CCK8, Transwell assay and Western blot were performed to determine the function of DCHS1 in UCEC. RESULTS DCHS1 is differentially expressed in many cancers and its expression is significantly associated with tumor prognosis and diagnosis. DCHS1 expression was significantly correlated with the infiltration of cancer-associated fibroblasts (CAFs), Endothelial cell (ECs), and Hematopoietic stem cell in most cancers. In addition, DCHS1 was significantly associated with sensitivity to many antitumor drugs. Functional enrichment analysis revealed that DCHS1-related proteins were involved in Focal adhesion, Endometrial cancer and Wnt signaling pathway. GSEA results showed that DCHS1 was related to epithelial-mesenchymal transition (EMT) in many cancers. In vitro experiments in UCEC showed that DCHS1 regulated cell proliferation, migration and EMT. CONCLUSIONS Our findings indicated that DCHS1 might be a novel prognostic and diagnostic biomarker and immunotherapy target, and plays an important role in the proliferation, migration and EMT in UCEC.
Collapse
Affiliation(s)
- Cai Meijuan
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), No.758 Hefei Road, Qingdao, 266035, Shandong, China
| | - Min Fang
- Department of Gynecology, Qingdao Women's and Children's Hospital, Qingdao University, Qingdao, Shandong, China
| | - Wang Qian
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), No.758 Hefei Road, Qingdao, 266035, Shandong, China.
| |
Collapse
|
28
|
Agorku DJ, Bosio A, Alves F, Ströbel P, Hardt O. Colorectal cancer-associated fibroblasts inhibit effector T cells via NECTIN2 signaling. Cancer Lett 2024; 595:216985. [PMID: 38821255 DOI: 10.1016/j.canlet.2024.216985] [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: 03/08/2024] [Revised: 04/29/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
Cancer-associated fibroblasts play a crucial role within the tumor microenvironment. However, a comprehensive characterization of CAF in colorectal cancer (CRC) is still missing. We combined scRNA-seq and spatial proteomics to decipher fibroblast heterogeneity in healthy human colon and CRC at high resolution. Analyzing nearly 23,000 fibroblasts, we identified 11 distinct clusters and verified them by spatial proteomics. Four clusters, consisting of myofibroblastic CAF (myCAF)-like, inflammatory CAF (iCAF)-like and proliferating fibroblasts as well as a novel cluster, which we named "T cell-inhibiting CAF" (TinCAF), were primarily found in CRC. This new cluster was characterized by the expression of immune-interacting receptors and ligands, including CD40 and NECTIN2. Co-culture of CAF and T cells resulted in a reduction of the effector T cell compartment, impaired proliferation, and increased exhaustion. By blocking its receptor interaction, we demonstrated that NECTIN2 was the key driver of T cell inhibition. Analysis of clinical datasets showed that NECTIN2 expression is a poor prognostic factor in CRC and other tumors. In conclusion, we identified a new class of immuno-suppressive CAF with features rendering them a potential target for future immunotherapies.
Collapse
Affiliation(s)
- David J Agorku
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany; University Medical Center Göttingen (UMG), Institute of Pathology, Göttingen, Lower Saxony, Germany
| | - Andreas Bosio
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Frauke Alves
- University Medical Center Göttingen, Department of Hematology and Medical Oncology, Göttingen, Lower Saxony, Germany; University Medical Center Göttingen, Institute for Diagnostic and Interventional Radiology, Göttingen, Lower Saxony, Germany; Max Planck Institute for Multidisciplinary Sciences, Translational Molecular Imaging, Göttingen, Lower Saxony, Germany
| | - Philipp Ströbel
- University Medical Center Göttingen (UMG), Institute of Pathology, Göttingen, Lower Saxony, Germany
| | - Olaf Hardt
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany.
| |
Collapse
|
29
|
Sajjadi SF, Salehi N, Sadeghi M. Comprehensive integrated single-cell RNA sequencing analysis of brain metastasis and glioma microenvironment: Contrasting heterogeneity landscapes. PLoS One 2024; 19:e0306220. [PMID: 39058687 PMCID: PMC11280140 DOI: 10.1371/journal.pone.0306220] [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: 01/31/2024] [Accepted: 06/12/2024] [Indexed: 07/28/2024] Open
Abstract
Understanding the specific type of brain malignancy, source of brain metastasis, and underlying transformation mechanisms can help provide better treatment and less harm to patients. The tumor microenvironment plays a fundamental role in cancer progression and affects both primary and metastatic cancers. The use of single-cell RNA sequencing to gain insights into the heterogeneity profiles in the microenvironment of brain malignancies is useful for guiding treatment decisions. To comprehensively investigate the heterogeneity in gliomas and brain metastasis originating from different sources (lung and breast), we integrated data from three groups of single-cell RNA-sequencing datasets obtained from GEO. We gathered and processed single-cell RNA sequencing data from 90,168 cells obtained from 17 patients. We then employed the R package Seurat for dataset integration. Next, we clustered the data within the UMAP space and acquired differentially expressed genes for cell categorization. Our results underscore the significance of macrophages as abundant and pivotal constituents of gliomas. In contrast, lung-to-brain metastases exhibit elevated numbers of AT2, cytotoxic CD4+ T, and exhausted CD8+ T cells. Conversely, breast-to-brain metastases are characterized by an abundance of epithelial and myCAF cells. Our study not only illuminates the variation in the TME between brain metastasis with different origins but also opens the door to utilizing established markers for these cell types to differentiate primary brain metastatic cancers.
Collapse
Affiliation(s)
- Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Najmeh Salehi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Mehdi Sadeghi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
- National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
30
|
Owaki T, Iida T, Miyai Y, Kato K, Hase T, Ishii M, Ando R, Hinohara K, Akashi T, Mizutani Y, Ishikawa T, Mii S, Shiraki Y, Esaki N, Yamamoto M, Tsukamoto T, Nomura S, Murakami T, Takahashi M, Yuguchi Y, Maeda M, Sano T, Sassa N, Matsukawa Y, Kawashima H, Akamatsu S, Enomoto A. Synthetic retinoid-mediated preconditioning of cancer-associated fibroblasts and macrophages improves cancer response to immune checkpoint blockade. Br J Cancer 2024; 131:372-386. [PMID: 38849479 PMCID: PMC11263587 DOI: 10.1038/s41416-024-02734-3] [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: 11/29/2023] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND The proliferation of cancer-associated fibroblasts (CAFs) hampers drug delivery and anti-tumor immunity, inducing tumor resistance to immune checkpoint blockade (ICB) therapy. However, it has remained a challenge to develop therapeutics that specifically target or modulate CAFs. METHODS We investigated the involvement of Meflin+ cancer-restraining CAFs (rCAFs) in ICB efficacy in patients with clear cell renal cell carcinoma (ccRCC) and urothelial carcinoma (UC). We examined the effects of Am80 (a synthetic retinoid) administration on CAF phenotype, the tumor immune microenvironment, and ICB efficacy in cancer mouse models. RESULTS High infiltration of Meflin+ CAFs correlated with ICB efficacy in patients with ccRCC and UC. Meflin+ CAF induction by Am80 administration improved ICB efficacy in the mouse models of cancer. Am80 exerted this effect when administered prior to, but not concomitant with, ICB therapy in wild-type but not Meflin-deficient mice. Am80-mediated induction of Meflin+ CAFs was associated with increases in antibody delivery and M1-like tumor-associated macrophage (TAM) infiltration. Finally, we showed the role of Chemerin produced from CAFs after Am80 administration in the induction of M1-like TAMs. CONCLUSION Our data suggested that Am80 administration prior to ICB therapy increases the number of Meflin+ rCAFs and ICB efficacy by inducing changes in TAM phenotype.
Collapse
Affiliation(s)
- Takayuki Owaki
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tadashi Iida
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Yuki Miyai
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Katsuhiro Kato
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsunari Hase
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryota Ando
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kunihiko Hinohara
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute for Advanced Research, Nagoya University, Nagoya, Japan
| | - Tomohiro Akashi
- Division of Systems Biology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Yasuyuki Mizutani
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takuya Ishikawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinji Mii
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Shiraki
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobutoshi Esaki
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masami Yamamoto
- Laboratory of Physiological Pathology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Tetsuya Tsukamoto
- Division of Analytical Pathology, Oncology Innovation Center, Fujita Health University, Toyoake, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
| | - Takashi Murakami
- Department of Microbiology, Saitama Medical University, Saitama, Japan
| | - Masahide Takahashi
- Department of Pathology, Fujita Health University, Toyoake, Japan
- International Center for Cell and Gene Therapy, Fujita Health University, Toyoake, Japan
| | - Yuri Yuguchi
- Department of Urology, Chukyo Hospital, Nagoya, Japan
| | | | - Tomoyasu Sano
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoto Sassa
- Department of Urology, Aichi Medical University, Nagakute, Japan
| | - Yoshihisa Matsukawa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shusuke Akamatsu
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu, Japan.
| |
Collapse
|
31
|
Dong Y, Bai J, Zhou J. Developing a dormancy-associated ECM signature in TNBC that is linked to immunosuppressive tumor microenvironment and selective sensitivity to MAPK inhibitors. Heliyon 2024; 10:e32106. [PMID: 38868025 PMCID: PMC11168407 DOI: 10.1016/j.heliyon.2024.e32106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/12/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024] Open
Abstract
Aims Cellular dormancy is a state of quiescence subpopulation of tumor cells, characterized by low differentiation and lack of mitotic activity. They could evade chemotherapy and targeted therapy, leading to drug resistance and disease recurrence. Recent studies have shown a correlation between dormant cancer cells and unique extracellular matrix (ECM) composition, which is critical in regulating cell behavior. However, their interacting roles in TNBC patients remains to be characterized. Main methods Dormant cancer cells in MDA-MB-231 cell line with highest PKH26 dye-retaining were FACS-sorted and gene expression was then analyzed. Dormant associated ECM (DA-ECM) signature was characterized by pathway analysis. Unsupervised hierarchical clustering was used to define distinct ECM features for TNBC patients. ECM-specific tumor biology was defined by integration of bulk RNA-seq with single-cell RNA-seq data, analysis of ligand-receptor interactions and enriched biological pathways, and in silico drug screening. We validated the sensitivity of dormant cancer cells to MAPK inhibitors by flow cytometry in vitro. Key findings We observed that dormant TNBC cells preferentially expressed ∼10 % DA-ECM genes. The DA-ECM High subtype defined by unsupervised hierarchical clustering analysis was associated with immunosuppressive tumor microenvironment. Moreover, ligand-receptor interaction and pathway analysis revealed that the DA-ECM High subtype may likely help maintain tumor cell dormancy through MAPK, Hedgehog and Notch signaling pathways. Finally, in silico drug screening against the DA-ECM signature and in vitro assay showed dormant cancer cells were relatively sensitive to the MAPK pathway inhibitors, which may represent a potential therapeutic strategy for treating TNBC. Significance Collectively, our research revealed that dormancy-associated ECM characterized tumor cells possess significant ECM remodeling capacity, and treatment strategies towards these cells could improve TNBC patient outcome.
Collapse
Affiliation(s)
- Yang Dong
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Jin Bai
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Jianjun Zhou
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| |
Collapse
|
32
|
Wu J, Lu Q, Zhao J, Wu W, Wang Z, Yu G, Tian G, Gao Z, Wang Q. Enhancing the Inhibition of Breast Cancer Growth Through Synergistic Modulation of the Tumor Microenvironment Using Combined Nano-Delivery Systems. Int J Nanomedicine 2024; 19:5125-5138. [PMID: 38855730 PMCID: PMC11162247 DOI: 10.2147/ijn.s460874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/17/2024] [Indexed: 06/11/2024] Open
Abstract
Purpose Breast cancer is a prevalent malignancy among women worldwide, and malignancy is closely linked to the tumor microenvironment (TME). Here, we prepared mixed nano-sized formulations composed of pH-sensitive liposomes (Ber/Ru486@CLPs) and small-sized nano-micelles (Dox@CLGs). These liposomes and nano-micelles were modified by chondroitin sulfate (CS) to selectively target breast cancer cells. Methods Ber/Ru486@CLPs and Dox@CLGs were prepared by thin-film dispersion and ethanol injection, respectively. To mimic actual TME, the in vitro "condition medium of fibroblasts + MCF-7" cell model and in vivo "4T1/NIH-3T3" co-implantation mice model were established to evaluate the anti-tumor effect of drugs. Results The physicochemical properties showed that Dox@CLGs and Ber/Ru486@CLPs were 28 nm and 100 nm in particle size, respectively. In vitro experiments showed that the mixed formulations significantly improved drug uptake and inhibited cell proliferation and migration. The in vivo anti-tumor studies further confirmed the enhanced anti-tumor capabilities of Dox@CLGs + Ber/Ru486@CLPs, including smaller tumor volumes, weak collagen deposition, and low expression levels of α-SMA and CD31 proteins, leading to a superior anti-tumor effect. Conclusion In brief, this combination therapy based on Dox@CLGs and Ber/Ru486@CLPs could effectively inhibit tumor development, which provides a promising approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Jingliang Wu
- School of Medicine, Weifang University of Science and Technology, Weifang, 262700, People’s Republic of China
| | - Qiao Lu
- School of Medicine, Weifang University of Science and Technology, Weifang, 262700, People’s Republic of China
- School of Life Science and Technology, Shandong Second Medical University, Weifang, 261053, People’s Republic of China
| | - Jialin Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261053, People’s Republic of China
| | - Wendi Wu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261053, People’s Republic of China
| | - Zhihua Wang
- School of Medicine, Weifang University of Science and Technology, Weifang, 262700, People’s Republic of China
| | - Guohua Yu
- Department of Oncology, Weifang People’s Hospital, Weifang, 261000, People’s Republic of China
| | - Guixiang Tian
- School of Life Science and Technology, Shandong Second Medical University, Weifang, 261053, People’s Republic of China
| | - Zhiqin Gao
- School of Life Science and Technology, Shandong Second Medical University, Weifang, 261053, People’s Republic of China
| | - Qing Wang
- Department of Stomatology, Weifang People’s Hospital, Weifang, 261000, People’s Republic of China
| |
Collapse
|
33
|
Qin Q, Yu R, Eriksson JE, Tsai HI, Zhu H. Cancer-associated fibroblasts in pancreatic ductal adenocarcinoma therapy: Challenges and opportunities. Cancer Lett 2024; 591:216859. [PMID: 38615928 DOI: 10.1016/j.canlet.2024.216859] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a solid organ malignancy with a high mortality rate. Statistics indicate that its incidence has been increasing as well as the associated deaths. Most patients with PDAC show poor response to therapies making the clinical management of this cancer difficult. Stromal cells in the tumor microenvironment (TME) contribute to the development of resistance to therapy in PDAC cancer cells. Cancer-associated fibroblasts (CAFs), the most prevalent stromal cells in the TME, promote a desmoplastic response, produce extracellular matrix proteins and cytokines, and directly influence the biological behavior of cancer cells. These multifaceted effects make it difficult to eradicate tumor cells from the body. As a result, CAF-targeting synergistic therapeutic strategies have gained increasing attention in recent years. However, due to the substantial heterogeneity in CAF origin, definition, and function, as well as high plasticity, majority of the available CAF-targeting therapeutic approaches are not effective, and in some cases, they exacerbate disease progression. This review primarily elucidates on the effect of CAFs on therapeutic efficiency of various treatment modalities, including chemotherapy, radiotherapy, immunotherapy, and targeted therapy. Strategies for CAF targeting therapies are also discussed.
Collapse
Affiliation(s)
- Qin Qin
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China
| | - Rong Yu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China
| | - John E Eriksson
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, FI-20520 Finland
| | - Hsiang-I Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
34
|
Banihashemian SS, Divband G, Pirayesh E, Nikkholgh B, Amini H, Shahrnoy AA, Nami R, Akbari ME. [ 68Ga]Ga-FAP-2286, a novel promising theragnostic approach for PET/CT imaging in patients with various type of metastatic cancers. Eur J Nucl Med Mol Imaging 2024; 51:1981-1988. [PMID: 38376804 DOI: 10.1007/s00259-024-06635-8] [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: 11/06/2023] [Accepted: 02/04/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND Fibroblast activation protein (FAP) has emerged as a promising target for diagnosis and therapeutic intervention due to high expression and accumulation in the stromal compartments of a variety of malignant tumors. FAP-2286 utilizes cyclic peptides with FAP-binding characteristics to enhance the retention of the imaging agent within tumors, in contrast to the small-molecule FAP inhibitors (FAPI) like FAPI-04/46. The aim of this study was to quantify the tumor uptake of [68Ga] Gallium-FAP-2286 within primary solid tumors, adjacent excised tissues, and metastatic lesions. METHODS In this prospective study, 21 patients (average age 51.9) with various diagnoses of remaining and metastatic cancers participated. Among them, six had metastatic sarcoma, and 14 had adenocarcinoma, including eight breast, two rectum, two lung, two pancreas, and one thyroid cases. The patients underwent a [68Ga]Ga-FAP-2286 PET/CT scan. An hour post-administration of [68Ga]Ga-FAP-2286, a visual assessment of whole body scans and semi-quantification of the PET/CT results were carried out. The standardized uptake values (SUV)max of [68Ga]Ga-FAP-2286 in tumor lesions and the tumor-to-background ratio (TBR) were then calculated. RESULTS The vital signs of the patients, such as heart rate, blood pressure, and temperature, were observed before, during, and after the diagnostic procedure during the 4-h follow-up. All individuals underwent the [68Ga]Ga-FAP-2286 PET/CT scans without any signs of drug-associated pharmacological effects. The PET/CT scans displayed substantial absorption of [68Ga]Ga-FAP-2286 in tumor lesions in all patients (100% (21/21)). Irrespective of the tumors' origins (epithelial or mesothelium) and whether they exhibited local recurrence, distant recurrence, or metastatic lesions, the PET/CT scans revealed the uptake of [68Ga]Ga-FAP-2286 in these lesions. CONCLUSION Overall, these data suggest that [68Ga]Ga-FAP-2286 is a promising FAP derivative for efficient metastatic cancer diagnosis and being considered as a potential compound for therapeutic application in patients with advanced metastatic cancers.
Collapse
Affiliation(s)
| | | | - Elahe Pirayesh
- Department of Nuclear Medicine, School of Medicine, Shohada'E Tajrish Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
35
|
Liu L, Xu L, Wu D, Zhu Y, Li X, Xu C, Chen K, Lin Y, Lao J, Cai P, Li X, Luo Y, Li X, Huang J, Lin T, Zhong W. Impact of tumour stroma-immune interactions on survival prognosis and response to neoadjuvant chemotherapy in bladder cancer. EBioMedicine 2024; 104:105152. [PMID: 38728838 PMCID: PMC11090066 DOI: 10.1016/j.ebiom.2024.105152] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND The tumour stroma is associated with unfavourable prognosis in diverse solid tumours, but its prognostic and predictive value in bladder cancer (BCa) is unclear. METHODS In this multicentre, retrospective study, we included 830 patients with BCa from six independent cohorts. Differences in overall survival (OS) and cancer-specific survival (CSS) were investigated between high-tumour stroma ratio (TSR) and low-TSR groups. Multi-omics analyses, including RNA sequencing, immunohistochemistry, and single-cell RNA sequencing, were performed to study stroma-immune interactions. TSR prediction models were developed based on pelvic CT scans, and the best performing model was selected based on receiver operator characteristic analysis. FINDINGS Compared to low-TSR tumours, high-TSR tumours were significantly associated with worse OS (HR = 1.193, 95% CI: 1.046-1.361, P = 0.008) and CSS (HR = 1.337, 95% CI: 1.139-1.569, P < 0.001), and lower rate of pathological complete response (pCR) to neoadjuvant chemotherapy (NAC). High-TSR tumours exhibited higher infiltration of immunosuppressive cells, including Tregs and tumour-associated neutrophils, while low-TSR tumours exhibited higher infiltration of immune-activating cells such as CD8+ Teff and XCR1+ dendritic cells. The TSR prediction model was developed by combining the intra-tumour and tumour base radiomics features, and showed good performance to predict high-TSR, as indicted by area under the curve of 0.871 (95% CI: 0.821-0.921), 0.821 (95% CI: 0.731-0.911), and 0.801 (95% CI: 0.737-0.865) in the training, internal validation, and external validation cohorts, respectively. In patients with low predicted TSR, 92.3% (12/13) achieved pCR, while only 35.3% (6/17) of patients with high predicted TSR achieved pCR. INTERPRETATION The tumour stroma was found to be significantly associated with clinical outcomes in patients with BCa as a result of tumour stroma-immune interactions. The radiomics prediction model provided non-invasive evaluation of TSR and was able to predict pCR in patients receiving NAC for BCa. FUNDING This work was supported by National Natural Science Foundation of China (Grant No. 82373254 and 81961128027), Guangdong Provincial Natural Science Foundation (Grant No. 2023A1515010258), Science and Technology Planning Project of Guangdong Province (Grant No. 2023B1212060013). Science and Technology Program of Guangzhou (SL2022A04J01754), Sun Yat-Sen Memorial Hospital Clinical Research 5010 Program (Grant No. SYS-5010Z-202401).
Collapse
Affiliation(s)
- Libo Liu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China
| | - Longhao Xu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China
| | - Daqin Wu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China
| | - Yingying Zhu
- Clinical Research Design Division, Clinical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Xiaoyang Li
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Chunru Xu
- Department of Urology, Peking University First Hospital, Beijing, PR China
| | - Ke Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China
| | - Yi Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China
| | - Jianwen Lao
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China
| | - Peicong Cai
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Beijing, PR China
| | - Yun Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Xiang Li
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China.
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China.
| | - Wenlong Zhong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China.
| |
Collapse
|
36
|
Milosevic V, Östman A. Interactions between cancer-associated fibroblasts and T-cells: functional crosstalk with targeting and biomarker potential. Ups J Med Sci 2024; 129:10710. [PMID: 38863724 PMCID: PMC11165253 DOI: 10.48101/ujms.v129.10710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 06/13/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous cell population recognized as a key component of the tumour microenvironment (TME). Cancer-associated fibroblasts are known to play an important role in maintaining and remodelling the extracellular matrix (ECM) in the tumour stroma, supporting cancer progression and inhibiting the immune system's response against cancer cells. This review aims to summarize the immunomodulatory roles of CAFs, particularly focussing on their T-cell suppressive effects. Cancer-associated fibroblasts have several ways by which they can affect the tumour's immune microenvironment (TIME). For example, their interactions with macrophages and dendritic cells (DCs) create an immunosuppressive milieu that can indirectly affect T-cell anticancer immunity and enable immune evasion. In addition, a number of recent studies have confirmed CAF-mediated direct suppressive effects on T-cell anticancer capacity through ECM remodelling, promoting the expression of immune checkpoints, cytokine secretion and the release of extracellular vesicles. The consequential impact of CAFs on T-cell function is then reflected in affecting T-cell proliferation and apoptosis, migration and infiltration, differentiation and exhaustion. Emerging evidence highlights the existence of specific CAF subsets with distinct capabilities to modulate the immune landscape of TME in various cancers, suggesting the possibility of their exploitation as possible prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Vladan Milosevic
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Arne Östman
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Kazakova AN, Lukina MM, Anufrieva KS, Bekbaeva IV, Ivanova OM, Shnaider PV, Slonov A, Arapidi GP, Shender VO. Exploring the diversity of cancer-associated fibroblasts: insights into mechanisms of drug resistance. Front Cell Dev Biol 2024; 12:1403122. [PMID: 38818409 PMCID: PMC11137237 DOI: 10.3389/fcell.2024.1403122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/22/2024] [Indexed: 06/01/2024] Open
Abstract
Introduction: Among the various stromal cell types within the tumor microenvironment, cancer-associated fibroblasts (CAFs) emerge as the predominant constituent, exhibiting a diverse array of oncogenic functions not intrinsic to normal fibroblasts. Their involvement spans across all stages of tumorigenesis, encompassing initiation, progression, and metastasis. Current understanding posits the coexistence of distinct subpopulations of CAFs within the tumor microenvironment across a spectrum of solid tumors, showcasing both pro- and antitumor activities. Recent advancements in single-cell transcriptomics have revolutionized our ability to meticulously dissect the heterogeneity inherent to CAF populations. Furthermore, accumulating evidence underscores the pivotal role of CAFs in conferring therapeutic resistance to tumors against various drug modalities. Consequently, efforts are underway to develop pharmacological agents specifically targeting CAFs. Methods: This review embarks on a comprehensive analysis, consolidating data from 36 independent single-cell RNA sequencing investigations spanning 17 distinct human malignant tumor types. Results: Our exploration centers on elucidating CAF population markers, discerning their prognostic relevance, delineating their functional contributions, and elucidating the underlying mechanisms orchestrating chemoresistance. Discussion: Finally, we deliberate on the therapeutic potential of harnessing CAFs as promising targets for intervention strategies in clinical oncology.
Collapse
Affiliation(s)
- Anastasia N. Kazakova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Maria M. Lukina
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - Ksenia S. Anufrieva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Irina V. Bekbaeva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
| | - Olga M. Ivanova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Polina V. Shnaider
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Andrey Slonov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Georgij P. Arapidi
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Victoria O. Shender
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
38
|
Kundu M, Butti R, Panda VK, Malhotra D, Das S, Mitra T, Kapse P, Gosavi SW, Kundu GC. Modulation of the tumor microenvironment and mechanism of immunotherapy-based drug resistance in breast cancer. Mol Cancer 2024; 23:92. [PMID: 38715072 PMCID: PMC11075356 DOI: 10.1186/s12943-024-01990-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 04/02/2024] [Indexed: 05/12/2024] Open
Abstract
Breast cancer, the most frequent female malignancy, is often curable when detected at an early stage. The treatment of metastatic breast cancer is more challenging and may be unresponsive to conventional therapy. Immunotherapy is crucial for treating metastatic breast cancer, but its resistance is a major limitation. The tumor microenvironment (TME) is vital in modulating the immunotherapy response. Various tumor microenvironmental components, such as cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs), are involved in TME modulation to cause immunotherapy resistance. This review highlights the role of stromal cells in modulating the breast tumor microenvironment, including the involvement of CAF-TAM interaction, alteration of tumor metabolism leading to immunotherapy failure, and other latest strategies, including high throughput genomic screening, single-cell and spatial omics techniques for identifying tumor immune genes regulating immunotherapy response. This review emphasizes the therapeutic approach to overcome breast cancer immune resistance through CAF reprogramming, modulation of TAM polarization, tumor metabolism, and genomic alterations.
Collapse
Affiliation(s)
- Moumita Kundu
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, 751024, India
- Department of Pharmaceutical Technology, Brainware University, West Bengal, 700125, India
| | - Ramesh Butti
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Venketesh K Panda
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, 751024, India
| | - Diksha Malhotra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, 751024, India
| | - Sumit Das
- National Centre for Cell Sciences, Savitribai Phule Pune University Campus, Pune, 411007, India
| | - Tandrima Mitra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, 751024, India
| | - Prachi Kapse
- School of Basic Medical Sciences, Savitribai Phule Pune University, Pune, 411007, India
| | - Suresh W Gosavi
- School of Basic Medical Sciences, Savitribai Phule Pune University, Pune, 411007, India
| | - Gopal C Kundu
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, 751024, India.
- Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to be University, Bhubaneswar, 751024, India.
| |
Collapse
|
39
|
You S, Han X, Xu Y, Sui L, Song K, Yao Q. High expression of SLC7A1 in high-grade serous ovarian cancer promotes tumor progression and is involved in MAPK/ERK pathway and EMT. Cancer Med 2024; 13:e7217. [PMID: 38752472 PMCID: PMC11097251 DOI: 10.1002/cam4.7217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/01/2024] [Accepted: 04/15/2024] [Indexed: 05/19/2024] Open
Abstract
Our previous studies have shown that upregulation of SLC7A1 in epithelial ovarian cancer (EOC) tumor cells significantly increases cancer cell proliferation, migration, and cisplatin resistance; however, the molecular mechanism by which SLC7A1 functions in EOC remains unknown. In later studies, we found that SLC7A1 is also highly expressed in the interstitial portion of high-grade serous ovarian cancer (HGSOC), but the significance of this high expression in the interstitial remains unclear. Here, we showed the Interstitial high expression of SLC7A1 in HGSOC by immunohistochemistry. SLC7A1 enriched in cancer-associated fibroblasts (CAFs) was upregulated by TGF-β1. Transwell assay, scratch assay, cck8 assay and cell adhesion assay showed that SLC7A1 highly expressed in CAFs promoted tumor cells invasion, migration and metastasis in vitro. The effect of SLC7A1 on MAPK and EMT pathway proteins in ovarian cancer (OC) was verified by RNA sequencing and western blotting. Overexpression of SLC7A1 in OC is involved in MAPK/ ERK pathway and EMT. In general, in HGSOC, CAFs overexpressing SLC7A1 supported the migration and invasion of tumor cells; SLC7A1 is highly expressed in ovarian cancer and is involved in ERK phosphorylation and EMT signaling in MAPK signaling pathway. This suggests that SLC7A1 may be a potential therapeutic target for OC metastasis.
Collapse
Affiliation(s)
- Shijing You
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Xiahui Han
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Yuance Xu
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Lei Sui
- Department of Gynecological OncologyAffiliated Qingdao Central Hospital of Qingdao UniversityQingdaoShandongChina
| | - Kejuan Song
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Qin Yao
- Department of Obstetrics and GynecologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| |
Collapse
|
40
|
Zhang X, Zhu R, Yu D, Wang J, Yan Y, Xu K. Single-cell RNA sequencing to explore cancer-associated fibroblasts heterogeneity: "Single" vision for "heterogeneous" environment. Cell Prolif 2024; 57:e13592. [PMID: 38158643 PMCID: PMC11056715 DOI: 10.1111/cpr.13592] [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: 07/18/2023] [Revised: 10/24/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs), a phenotypically and functionally heterogeneous stromal cell, are one of the most important components of the tumour microenvironment. Previous studies have consolidated it as a promising target against cancer. However, variable therapeutic efficacy-both protumor and antitumor effects have been observed not least owing to the strong heterogeneity of CAFs. Over the past 10 years, advances in single-cell RNA sequencing (scRNA-seq) technologies had a dramatic effect on biomedical research, enabling the analysis of single cell transcriptomes with unprecedented resolution and throughput. Specifically, scRNA-seq facilitates our understanding of the complexity and heterogeneity of diverse CAF subtypes. In this review, we discuss the up-to-date knowledge about CAF heterogeneity with a focus on scRNA-seq perspective to investigate the emerging strategies for integrating multimodal single-cell platforms. Furthermore, we summarized the clinical application of scRNA-seq on CAF research. We believe that the comprehensive understanding of the heterogeneity of CAFs form different visions will generate innovative solutions to cancer therapy and achieve clinical applications.
Collapse
Affiliation(s)
- Xiangjian Zhang
- The Dingli Clinical College of Wenzhou Medical UniversityWenzhouZhejiangChina
- Department of Surgical OncologyWenzhou Central HospitalWenzhouZhejiangChina
- The Second Affiliated Hospital of Shanghai UniversityWenzhouZhejiangChina
| | - Ruiqiu Zhu
- Interventional Cancer Institute of Chinese Integrative MedicinePutuo Hospital, Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Die Yu
- Interventional Cancer Institute of Chinese Integrative MedicinePutuo Hospital, Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Juan Wang
- School of MedicineShanghai UniversityShanghaiChina
| | - Yuxiang Yan
- The Dingli Clinical College of Wenzhou Medical UniversityWenzhouZhejiangChina
- Department of Surgical OncologyWenzhou Central HospitalWenzhouZhejiangChina
- The Second Affiliated Hospital of Shanghai UniversityWenzhouZhejiangChina
| | - Ke Xu
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
- Wenzhou Institute of Shanghai UniversityWenzhouChina
| |
Collapse
|
41
|
Mathieson L, Koppensteiner L, Dorward DA, O'Connor RA, Akram AR. Cancer-associated fibroblasts expressing fibroblast activation protein and podoplanin in non-small cell lung cancer predict poor clinical outcome. Br J Cancer 2024; 130:1758-1769. [PMID: 38582812 PMCID: PMC11130154 DOI: 10.1038/s41416-024-02671-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are a dominant cell type in the stroma of non-small cell lung cancer (NSCLC). Fibroblast heterogeneity reflects subpopulations of CAFs, which can influence prognosis and treatment efficacy. We describe the subtypes of CAFs in NSCLC. METHODS Primary human NSCLC resections were assessed by flow cytometry and multiplex immunofluorescence for markers of fibroblast activation which allowed identification of CAF subsets. Survival data were analysed for our NSCLC cohort consisting of 163 patients to understand prognostic significance of CAF subsets. RESULTS We identified five CAF populations, termed CAF S1-S5. CAF-S5 represents a previously undescribed population, and express FAP and PDPN but lack the myofibroblast marker αSMA, whereas CAF-S1 populations express all three. CAF-S5 are spatially further from tumour regions then CAF-S1 and scRNA data demonstrate an inflammatory phenotype. The presence of CAF-S1 or CAF-S5 is correlated to worse survival outcome in NSCLC, despite curative resection, highlighting the prognostic importance of CAF subtypes in NSCLC. TCGA data suggest the predominance of CAF-S5 has a poor prognosis across several cancer types. CONCLUSION This study describes the fibroblast heterogeneity in NSCLC and the prognostic importance of the novel CAF-S5 subset where its presence correlates to worse survival outcome.
Collapse
Affiliation(s)
- Layla Mathieson
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Lilian Koppensteiner
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - David A Dorward
- Department of Pathology, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Richard A O'Connor
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Ahsan R Akram
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK.
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK.
- Cancer Research UK Scotland Centre, Institute of Genetics & Cancer, The University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK.
| |
Collapse
|
42
|
Xie Z, Qin Y, Chen X, Yang S, Yang J, Gui L, Liu P, He X, Zhou S, Zhang C, Tang L, Shi Y. Deciphering the Prognostic Significance of MYD88 and CD79B Mutations in Diffuse Large B-Cell Lymphoma: Insights into Treatment Outcomes. Target Oncol 2024; 19:383-400. [PMID: 38643457 DOI: 10.1007/s11523-024-01057-w] [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] [Accepted: 03/15/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND The clinical and genetic characteristics, as well as treatment outcomes, of diffuse large B-cell lymphoma (DLBCL) patients with different MYD88 and CD79B mutation status merit further investigation. OBJECTIVE This study aims to investigate the distinctions in clinical manifestations, genetic characteristics, and treatment outcomes among MYD88-CD79Bco-mut, MYD88/CD79Bsingle-mut, and MYD88-CD79Bco-wt DLBCL patients. PATIENTS AND METHODS Clinical and genetic characteristics, along with treatment outcomes among 2696 DLBCL patients bearing MYD88-CD79Bco-mut, MYD88/CD79Bsingle-mut, and MYD88-CD79Bco-wt treated with R-CHOP/R-CHOP-like regimens from the Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College and six external cohorts were analyzed. Potential molecular mechanisms were investigated through Gene Set Enrichment Analysis and xCell methodology. RESULTS In the MCD subtype, patients with MYD88-CD79Bco-mut showed comparable progression-free survival (PFS) and overall survival (OS) compared to MYD88/CD79Bsingle-mut or MYD88-CD79Bco-wt. However, in the non-MCD subtype, patients with MYD88-CD79Bco-mut exhibited significantly inferior OS than MYD88/CD79Bsingle-mut or MYD88-CD79Bco-wt, while there was no significant OS difference between MYD88/CD79Bsingle-mut and MYD88-CD79Bco-wt (median OS: 68.8 [95% CI 22-NA] vs NA [95% CI 112-NA] vs 177.7 [95% CI 159-NA] months; MYD88-CD79Bco-mut vs MYD88/CD79Bsingle-mut: p = 0.02; MYD88-CD79Bco-mut vs MYD88-CD79Bco-wt: p = 0.03; MYD88/CD79Bsingle-mut vs MYD88-CD79Bco-wt: p = 0.33). Regarding patients with MYD88-CD79Bco-mut, there was no significant difference in PFS and OS between the MCD and non-MCD subtypes. Within the MYD88-CD79Bco-mut group, patients with PIM1mut had better PFS than PIM1wt (median PFS: 8.34 [95% CI 5.56-NA] vs 43.8 [95% CI 26.4-NA] months; p = 0.02). Possible mechanisms contributing to the superior PFS of PIM1mut patients may include activated lymphocyte-mediated immunity and interferon response, a higher proportion of natural killer T cells and plasmacytoid dendritic cells, as well as suppressed angiogenesis and epithelial-mesenchymal transition, along with lower fibroblast and stromal score. CONCLUSIONS In the MCD subtype, patients with MYD88-CD79Bco-mut showed comparable PFS and OS compared to MYD88/CD79Bsingle-mut or MYD88-CD79Bco-wt, while in the non-MCD subtype, they exhibited significantly inferior OS. There was no significant disparity in PFS and OS of MYD88-CD79Bco-mut between the MCD and non-MCD subtypes. The presence of PIM1mut within the MYD88-CD79Bco-mut group correlated with better PFS, which may result from an intricate interplay of immune processes and tumor microenvironment alterations.
Collapse
Affiliation(s)
- Zucheng Xie
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Yan Qin
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Xinrui Chen
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Sheng Yang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Jianliang Yang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Lin Gui
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Peng Liu
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Xiaohui He
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Shengyu Zhou
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Changgong Zhang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Le Tang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Yuankai Shi
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
43
|
Lou H, Lin X, Wei G, Wu Z, Xiao Y. Construction of an Anoikis-Related Gene Prognostic Signature and Identification of ANGPTL4 as a Key Oncogene in Lung Adenocarcinoma. Mol Biotechnol 2024; 66:1290-1302. [PMID: 38381376 DOI: 10.1007/s12033-023-01031-3] [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: 09/15/2023] [Accepted: 12/11/2023] [Indexed: 02/22/2024]
Abstract
Anoikis plays an important role in cancer invasion and metastasis. However, the role of anoikis-related genes, AnRGs, in lung adenocarcinoma (LUAD) is not clear. First, anoikis-related genes (AnRGs) were obtained from the Genecard database. Second, the prognostic risk model of AnRGs was established by univariate Cox analysis, the Least Absolute Shrinkage and Selection Operator (LASSO) analysis, and multivariate Cox analysis. Finally, in vitro cell experiments were carried out to determine the expression and function of the key gene AnRGs. Three AnRGs (angiopoietin-like 4, ANGPTL4; Cyclin-Dependent Kinase Inhibitor 3, CDKN3; Solute Carrier Organic Anion Transporter Family Member 1B3, SLCO1B3) were screened for the construction of risk prediction model. Additionally, ANGPTL4 was significantly highly expressed in tumor cells, and the knockdown of ANGPTL4 expression on tumor cells could inhibit tumor cell migration and apoptosis. Constructing a risk model based on anoikis-related genes can effectively differentiate the prognosis of LUAD. ANGPTL4 can be used as a potential new target for LUAD treatment.
Collapse
Affiliation(s)
- Hao Lou
- School of Medicine, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Xuelian Lin
- Department of Oncology, Taikang Tongji (Wuhan) Hospital, Wuhan, People's Republic of China
| | - Guangyou Wei
- School of Medicine, Anhui University of Science and Technology, Huainan, People's Republic of China.
- Bo Zhou Municipal People's Hospital, Bo Zhou, People's Republic of China.
- Bozhou Clinical Medicine of Anhui University of Science & Technology, Bo Zhou, People's Republic of China.
| | - Zelai Wu
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, People's Republic of China
| | - Youde Xiao
- Department of Oncology, Taikang Tongji (Wuhan) Hospital, Wuhan, People's Republic of China.
| |
Collapse
|
44
|
Lee MR, Woo SM, Kim MK, Han S, Park S, Lee WJ, Lee D, Choi SI, Choi W, Yoon K, Chun JW, Kim Y, Kong S. Application of plasma circulating KRAS mutations as a predictive biomarker for targeted treatment of pancreatic cancer. Cancer Sci 2024; 115:1283-1295. [PMID: 38348576 PMCID: PMC11007020 DOI: 10.1111/cas.16104] [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: 09/12/2023] [Revised: 01/11/2024] [Accepted: 01/27/2024] [Indexed: 04/12/2024] Open
Abstract
Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations in circulating tumor deoxyribonucleic acid (ctDNA) have been reported as representative noninvasive prognostic markers for pancreatic ductal adenocarcinoma (PDAC). Here, we aimed to evaluate single KRAS mutations as prognostic and predictive biomarkers, with an emphasis on potential therapeutic approaches to PDAC. A total of 128 patients were analyzed for multiple or single KRAS mutations (G12A, G12C, G12D, G12R, G12S, G12V, and G13D) in their tumors and plasma using droplet digital polymerase chain reaction (ddPCR). Overall, KRAS mutations were detected by multiplex ddPCR in 119 (93%) of tumor DNA and 68 (53.1%) of ctDNA, with a concordance rate of 80% between plasma ctDNA and tumor DNA in the metastatic stage, which was higher than the 44% in the resectable stage. Moreover, the prognostic prediction of both overall survival (OS) and progression-free survival (PFS) was more relevant using plasma ctDNA than tumor DNA. Further, we evaluated the selective tumor-suppressive efficacy of the KRAS G12C inhibitor sotorasib in a patient-derived organoid (PDO) from a KRAS G12C-mutated patient using a patient-derived xenograft (PDX) model. Sotorasib showed selective inhibition in vitro and in vivo with altered tumor microenvironment, including fibroblasts and macrophages. Collectively, screening for KRAS single mutations in plasma ctDNA and the use of preclinical models of PDO and PDX with genetic mutations would impact precision medicine in the context of PDAC.
Collapse
Affiliation(s)
- Mi Rim Lee
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Molecular Imaging Branch, Division of Convergence TechnologyResearch Institute of National Cancer CenterGoyangKorea
| | - Sang Myung Woo
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer CenterGoyangKorea
- Immuno‐Oncology Branch, Division of Rare and Refractory CenterResearch Institute of National Cancer CenterGoyangKorea
| | - Min Kyeong Kim
- Targeted Therapy Branch, Division of Rare and Refractory CenterResearch Institute of National Cancer CenterGoyangKorea
| | - Sung‐Sik Han
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer CenterGoyangKorea
| | - Sang‐Jae Park
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer CenterGoyangKorea
| | - Woo Jin Lee
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer CenterGoyangKorea
- Interventional Medicine Branch, Division of Clinical ResearchResearch Institute of National Cancer CenterGoyangKorea
| | - Dong‐eun Lee
- Biostatistics Collaboration TeamResearch Core Center, National Cancer CenterGoyangKorea
| | - Sun Il Choi
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Molecular Imaging Branch, Division of Convergence TechnologyResearch Institute of National Cancer CenterGoyangKorea
- Henan Key Laboratory of Brain Targeted Bio‐Nanomedicine, School of Life Sciences & School of PharmacyHenan UniversityKaifengHenanChina
| | - Wonyoung Choi
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Center for Clinical Trials, Hospital, National Cancer CenterGoyangKorea
- Cancer Molecular Biology Branch, Division of Cancer BiologyResearch Institute of National Cancer CenterGoyangKorea
| | - Kyong‐Ah Yoon
- College of Veterinary MedicineKonkuk UniversitySeoulKorea
| | - Jung Won Chun
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer CenterGoyangKorea
- Interventional Medicine Branch, Division of Clinical ResearchResearch Institute of National Cancer CenterGoyangKorea
| | - Yun‐Hee Kim
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Molecular Imaging Branch, Division of Convergence TechnologyResearch Institute of National Cancer CenterGoyangKorea
| | - Sun‐Young Kong
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Targeted Therapy Branch, Division of Rare and Refractory CenterResearch Institute of National Cancer CenterGoyangKorea
- Department of Laboratory MedicineHospital, National Cancer CenterGoyangKorea
| |
Collapse
|
45
|
Lin Z, Yang L. Identification of a CpG-based signature coupled with gene expression as prognostic indicators for melanoma: a preliminary study. Sci Rep 2024; 14:5302. [PMID: 38438381 PMCID: PMC10912562 DOI: 10.1038/s41598-023-50614-2] [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: 02/20/2023] [Accepted: 12/22/2023] [Indexed: 03/06/2024] Open
Abstract
DNA methylation is an important part of the genomic biology, which recently allowed the identification of key biomarkers for a variety of cancers, including cutaneous melanoma. Despite the current knowledge in cutaneous melanoma, there is a clear need for new efficient biomarkers in clinical application of detection. We use The Cancer Genome Atlas data as a training set and a multi-stage screening strategy to identify prognostic characteristics of melanoma based on DNA methylation. Three DNA methylation CpG sites were identified to be related to the overall survival in the skin cutaneous melanoma cohort. This signature was validated in two independent datasets from Gene Expression Omnibus. The stratified analysis by clinical stage, age, gender, and grade retained the statistical significance. The methylation signature was significantly correlated with immune cells and anti-tumor immune response. Moreover, gene expression corresponding to the candidate CpG locus was also significantly correlated with the survival rate of the patient. About 49% of the prognostic effects of methylation are mediated by affecting the expression of the corresponding genes. The prognostic characteristics of DNA methylation combined with clinical information provide a better prediction value tool for melanoma patients than the clinical information alone. However, more experiments are required to validate these findings. Overall, this signature presents a prospect of novel and wide-ranging applications for appropriate clinical adjuvant trails.
Collapse
Affiliation(s)
- Zhen Lin
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Liu Yang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
46
|
Di Modugno F, Di Carlo A, Spada S, Palermo B, D'Ambrosio L, D'Andrea D, Morello G, Belmonte B, Sperduti I, Balzano V, Gallo E, Melchionna R, Panetta M, Campo G, De Nicola F, Goeman F, Antoniani B, Carpano S, Frigè G, Warren S, Gallina F, Lambrechts D, Xiong J, Vincent BG, Wheeler N, Bortone DS, Cappuzzo F, Facciolo F, Tripodo C, Visca P, Nisticò P. Tumoral and stromal hMENA isoforms impact tertiary lymphoid structure localization in lung cancer and predict immune checkpoint blockade response in patients with cancer. EBioMedicine 2024; 101:105003. [PMID: 38340557 PMCID: PMC10869748 DOI: 10.1016/j.ebiom.2024.105003] [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: 07/27/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Tertiary Lymphoid Structures (TLS) correlate with positive outcomes in patients with NSCLC and the efficacy of immune checkpoint blockade (ICB) in cancer. The actin regulatory protein hMENA undergoes tissue-specific splicing, producing the epithelial hMENA11a linked to favorable prognosis in early NSCLC, and the mesenchymal hMENAΔv6 found in invasive cancer cells and pro-tumoral cancer-associated fibroblasts (CAFs). This study investigates how hMENA isoforms in tumor cells and CAFs relate to TLS presence, localization and impact on patient outcomes and ICB response. METHODS Methods involved RNA-SEQ on NSCLC cells with depleted hMENA isoforms. A retrospective observational study assessed tissues from surgically treated N0 patients with NSCLC, using immunohistochemistry for tumoral and stromal hMENA isoforms, fibronectin, and TLS presence. ICB-treated patient tumors were analyzed using Nanostring nCounter and GeoMx spatial transcriptomics. Multiparametric flow cytometry characterized B cells and tissue-resident memory T cells (TRM). Survival and ICB response were estimated in the cohort and validated using bioinformatics pipelines in different datasets. FINDINGS Findings indicate that hMENA11a in NSCLC cells upregulates the TLS regulator LTβR, decreases fibronectin, and favors CXCL13 production by TRM. Conversely, hMENAΔv6 in CAFs inhibits LTβR-related NF-kB pathway, reduces CXCL13 secretion, and promotes fibronectin production. These patterns are validated in N0 NSCLC tumors, where hMENA11ahigh expression, CAF hMENAΔv6low, and stromal fibronectinlow are associated with intratumoral TLS, linked to memory B cells and predictive of longer survival. The hMENA isoform pattern, fibronectin, and LTβR expression broadly predict ICB response in tumors where TLS indicates an anti-tumor immune response. INTERPRETATION This study uncovers hMENA alternative splicing as an unexplored contributor to TLS-related Tumor Immune Microenvironment (TIME) and a promising biomarker for clinical outcomes and likely ICB responsiveness in N0 patients with NSCLC. FUNDING This work is supported by AIRC (IG 19822), ACC (RCR-2019-23669120), CAL.HUB.RIA Ministero Salute PNRR-POS T4, "Ricerca Corrente" granted by the Italian Ministry of Health.
Collapse
Affiliation(s)
- Francesca Di Modugno
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy.
| | - Anna Di Carlo
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Sheila Spada
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Belinda Palermo
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Lorenzo D'Ambrosio
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Daniel D'Andrea
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, New Hall Block - Room 171, Clifton Campus - NG11 8NS, Nottingham, United Kingdom
| | - Gaia Morello
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Corso Tukory 211, 90134, Palermo, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Corso Tukory 211, 90134, Palermo, Italy
| | - Isabella Sperduti
- Biostatistics and Scientific Direction, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Vittoria Balzano
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Enzo Gallo
- Pathology Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Roberta Melchionna
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Mariangela Panetta
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Giulia Campo
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Francesca De Nicola
- SAFU Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Frauke Goeman
- SAFU Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Barbara Antoniani
- Pathology Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Silvia Carpano
- Second Division of Medical Oncology, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Gianmaria Frigè
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, Milan, Italy
| | - Sarah Warren
- NanoString Technologies Inc., 530 Fairview Ave N, Seattle, WA, 98109, USA
| | - Filippo Gallina
- Thoracic-Surgery Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144 Rome, Italy
| | - Diether Lambrechts
- Center for Cancer Biology, Herestraat 49 box 912, VIB, 3000, Leuven, Belgium
| | - Jieyi Xiong
- Center for Cancer Biology, Herestraat 49 box 912, VIB, 3000, Leuven, Belgium
| | - Benjamin G Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 5206 Marsico Hall, Chapel Hill, NC, 27599, USA
| | - Nathan Wheeler
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 5206 Marsico Hall, Chapel Hill, NC, 27599, USA
| | - Dante S Bortone
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 5206 Marsico Hall, Chapel Hill, NC, 27599, USA
| | - Federico Cappuzzo
- Second Division of Medical Oncology, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Francesco Facciolo
- Thoracic-Surgery Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144 Rome, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Corso Tukory 211, 90134, Palermo, Italy
| | - Paolo Visca
- Pathology Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy.
| |
Collapse
|
47
|
Honda CK, Kurozumi S, Fujii T, Pourquier D, Khellaf L, Boissiere F, Horiguchi J, Oyama T, Shirabe K, Colinge J, Yokobori T, Turtoi A. Cancer-associated fibroblast spatial heterogeneity and EMILIN1 expression in the tumor microenvironment modulate TGF-β activity and CD8 + T-cell infiltration in breast cancer. Theranostics 2024; 14:1873-1885. [PMID: 38505604 PMCID: PMC10945331 DOI: 10.7150/thno.90627] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/13/2024] [Indexed: 03/21/2024] Open
Abstract
Rationale: The tumor microenvironment (TME) and its multifaceted interactions with cancer cells are major targets for cancer treatment. Single-cell technologies have brought major insights into the TME, but the resulting complexity often precludes conclusions on function. Methods: We combined single-cell RNA sequencing and spatial transcriptomic data to explore the relationship between different cancer-associated fibroblast (CAF) populations and immune cell exclusion in breast tumors. The significance of the findings was then evaluated in a cohort of tumors (N=75) from breast cancer patients using immunohistochemistry analysis. Results: Our data show for the first time the degree of spatial organization of different CAF populations in breast cancer. We found that IL-iCAFs, Detox-iCAFs, and IFNγ-iCAFs tended to cluster together, while Wound-myCAFs, TGFβ-myCAFs, and ECM-myCAFs formed another group that overlapped with elevated TGF-β signaling. Differential gene expression analysis of areas with CD8+ T-cell infiltration/exclusion within the TGF-β signaling-rich zones identified elastin microfibrillar interface protein 1 (EMILIN1) as a top modulated gene. EMILIN1, a TGF-β inhibitor, was upregulated in IFNγ-iCAFs directly modulating TGFβ immunosuppressive function. Histological analysis of 75 breast cancer samples confirmed that high EMILIN1 expression in the tumor margins was related to high CD8+ T-cell infiltration, consistent with our spatial gene expression analysis. High EMILIN1 expression was also associated with better prognosis of patients with breast cancer, underscoring its functional significance for the recruitment of cytotoxic T cells into the tumor area. Conclusion: Our data show that correlating TGF-β signaling to a CAF subpopulation is not enough because proteins with TGF-β-modulating activity originating from other CAF subpopulations can alter its activity. Therefore, therapeutic targeting should remain focused on biological processes rather than on specific CAF subtypes.
Collapse
Affiliation(s)
- Chikako Kanno Honda
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Sasagu Kurozumi
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- Department of Breast Surgery, International University of Health and Welfare, Chiba, Japan
| | - Takaaki Fujii
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Didier Pourquier
- Institut régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
- Tumor Microenvironment and Resistance to Treatment Lab, INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Lakhdar Khellaf
- Institut régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
- Tumor Microenvironment and Resistance to Treatment Lab, INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Florence Boissiere
- Institut régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Jun Horiguchi
- Department of Breast Surgery, International University of Health and Welfare, Chiba, Japan
| | - Tetsunari Oyama
- Department of Pathology and Diagnostics, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Ken Shirabe
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Jacques Colinge
- Institut régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
- Université de Montpellier, Montpellier, France
- Cancer Bioinformatics and Systems Biology Team, INSERM U1194, Montpellier, France
| | - Takehiko Yokobori
- Division of Integrated Oncology Research, Gunma University, Initiative for Advanced Research (GIAR), Maebashi, Gunma, Japan
| | - Andrei Turtoi
- Institut régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
- Tumor Microenvironment and Resistance to Treatment Lab, INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Division of Integrated Oncology Research, Gunma University, Initiative for Advanced Research (GIAR), Maebashi, Gunma, Japan
| |
Collapse
|
48
|
Zhang H, Lu X, Lu B, Gullo G, Chen L. Measuring the composition of the tumor microenvironment with transcriptome analysis: past, present and future. Future Oncol 2024; 20:1207-1220. [PMID: 38362731 PMCID: PMC11318690 DOI: 10.2217/fon-2023-0658] [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: 07/31/2023] [Accepted: 01/24/2024] [Indexed: 02/17/2024] Open
Abstract
Interactions between tumor cells and immune cells in the tumor microenvironment (TME) play a vital role the mechanisms of immune evasion, by which cancer cells escape immune elimination. Thus, the characterization and quantification of different components in the TME is a hot topic in molecular biology and drug discovery. Since the development of transcriptome sequencing in bulk tissue, single cells and spatial dimensions, there are increasing methods emerging to deconvolute and subtype the TME. This review discusses and compares such computational strategies and downstream subtyping analyses. Integrative analyses of the transcriptome with other data, such as epigenetics and T-cell receptor sequencing, are needed to obtain comprehensive knowledge of the dynamic TME.
Collapse
Affiliation(s)
- Han Zhang
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Xinghua Lu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Binfeng Lu
- Center for Discovery & Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Giuseppe Gullo
- Department of Obstetrics & Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146, Palermo, Italy
| | - Lujia Chen
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| |
Collapse
|
49
|
Licaj M, Mhaidly R, Kieffer Y, Croizer H, Bonneau C, Meng A, Djerroudi L, Mujangi-Ebeka K, Hocine HR, Bourachot B, Magagna I, Leclere R, Guyonnet L, Bohec M, Guérin C, Baulande S, Kamal M, Le Tourneau C, Lecuru F, Becette V, Rouzier R, Vincent-Salomon A, Gentric G, Mechta-Grigoriou F. Residual ANTXR1+ myofibroblasts after chemotherapy inhibit anti-tumor immunity via YAP1 signaling pathway. Nat Commun 2024; 15:1312. [PMID: 38346978 PMCID: PMC10861537 DOI: 10.1038/s41467-024-45595-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 01/29/2024] [Indexed: 02/15/2024] Open
Abstract
Although cancer-associated fibroblast (CAF) heterogeneity is well-established, the impact of chemotherapy on CAF populations remains poorly understood. Here we address this question in high-grade serous ovarian cancer (HGSOC), in which we previously identified 4 CAF populations. While the global content in stroma increases in HGSOC after chemotherapy, the proportion of FAP+ CAF (also called CAF-S1) decreases. Still, maintenance of high residual CAF-S1 content after chemotherapy is associated with reduced CD8+ T lymphocyte density and poor patient prognosis, emphasizing the importance of CAF-S1 reduction upon treatment. Single cell analysis, spatial transcriptomics and immunohistochemistry reveal that the content in the ECM-producing ANTXR1+ CAF-S1 cluster (ECM-myCAF) is the most affected by chemotherapy. Moreover, functional assays demonstrate that ECM-myCAF isolated from HGSOC reduce CD8+ T-cell cytotoxicity through a Yes Associated Protein 1 (YAP1)-dependent mechanism. Thus, efficient inhibition after treatment of YAP1-signaling pathway in the ECM-myCAF cluster could enhance CD8+ T-cell cytotoxicity. Altogether, these data pave the way for therapy targeting YAP1 in ECM-myCAF in HGSOC.
Collapse
Affiliation(s)
- Monika Licaj
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Rana Mhaidly
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Yann Kieffer
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Hugo Croizer
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Claire Bonneau
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
- Department of Surgery, Institut Curie Hospital Group, 35 rue Dailly, 92210, Saint-Cloud, France
| | - Arnaud Meng
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Lounes Djerroudi
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 26, rue d'Ulm, F-75248, Paris, France
| | - Kevin Mujangi-Ebeka
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Hocine R Hocine
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Brigitte Bourachot
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Ilaria Magagna
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France
| | - Renaud Leclere
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 26, rue d'Ulm, F-75248, Paris, France
| | - Lea Guyonnet
- Cytometry platform, PSL University, Institut Curie, 75005, Paris, France
| | - Mylene Bohec
- ICGex Next-Generation Sequencing Platform, PSL University, Institut Curie, 75005, Paris, France
| | - Coralie Guérin
- Cytometry platform, PSL University, Institut Curie, 75005, Paris, France
| | - Sylvain Baulande
- ICGex Next-Generation Sequencing Platform, PSL University, Institut Curie, 75005, Paris, France
| | - Maud Kamal
- Department of Drug Development and Innovation, Institut Curie Hospital Group, 26, rue d'Ulm, F-75248, Paris, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation, Institut Curie Hospital Group, 26, rue d'Ulm, F-75248, Paris, France
- INSERM, U900, Paris-Saclay University, Institut Curie, 35 rue Dailly, 92210, Saint-Cloud, France
| | - Fabrice Lecuru
- Breast, gynecology and reconstructive surgery Department, Institut Curie Hospital Group, Paris Cité University, 26, rue d'Ulm, F-75248, Paris, France
| | - Véronique Becette
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 35 rue Dailly, 92210, Saint-Cloud, France
| | - Roman Rouzier
- Department of Surgery, Institut Curie Hospital Group, 35 rue Dailly, 92210, Saint-Cloud, France
| | - Anne Vincent-Salomon
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 26, rue d'Ulm, F-75248, Paris, France
| | - Geraldine Gentric
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France.
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France.
| | - Fatima Mechta-Grigoriou
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75248, Paris, France.
- Inserm, U830, 26, rue d'Ulm, Paris, F-75005, France.
| |
Collapse
|
50
|
Yin SY, Liu YJ, Li JP, Liu J. Overexpression of FERM Domain Containing Kindlin 2 (FERMT2) in Fibroblasts Correlates with EMT and Immunosuppression in Gastric Cancer. Int J Genomics 2024; 2024:4123737. [PMID: 38352691 PMCID: PMC10864055 DOI: 10.1155/2024/4123737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/26/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024] Open
Abstract
The mesenchymal feature, dominated by epithelial mesenchymal transition (EMT) and stromal cell activation, is one of the main reasons for the aggressive nature of tumors, yet it remains poorly understood. In gastric cancer (GC), the fermitin family homolog-2 (FERMT2) is involved in macrophage signaling, promoting migration and invasion. However, the function of FERMT2 in fibroblasts remains unclear. Here, we demonstrated that downregulation of FERMT2 expression can block EMT in GC cells by inhibiting fibroblast activation in vitro. Furthermore, we found that, in addition to the known pathways, fibroblast-derived FERMT2 promotes M2-like macrophage growth and that in human GC samples, there is a strong positive correlation between FERMT2 and CD163 and CD206 levels. Notably, high FERMT2 expression was significantly associated with poor clinical outcomes and was upregulated in patients with advanced disease. Taken together, our results provide evidence that the fibroblast-FERMT2-EMT-M2 macrophage axis plays a critical role in the GC mesenchymal phenotype and may be a promising target for the treatment of advanced GC.
Collapse
Affiliation(s)
- Sheng-yan Yin
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Second Chinese Medicine Hospital, Nanjing, Jiangsu 210029, China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Yuan-jie Liu
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Second Chinese Medicine Hospital, Nanjing, Jiangsu 210029, China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Jie-pin Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Jian Liu
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Second Chinese Medicine Hospital, Nanjing, Jiangsu 210029, China
| |
Collapse
|