1
|
Xiao Z, Puré E. The fibroinflammatory response in cancer. Nat Rev Cancer 2025; 25:399-425. [PMID: 40097577 DOI: 10.1038/s41568-025-00798-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 03/19/2025]
Abstract
Fibroinflammation refers to the highly integrated fibrogenic and inflammatory responses mediated by the concerted function of fibroblasts and innate immune cells in response to tissue perturbation. This process underlies the desmoplastic remodelling of the tumour microenvironment and thus plays an important role in tumour initiation, growth and metastasis. More specifically, fibroinflammation alters the biochemical and biomechanical signalling in malignant cells to promote their proliferation and survival and further supports an immunosuppressive microenvironment by polarizing the immune status of tumours. Additionally, the presence of fibroinflammation is often associated with therapeutic resistance. As such, there is increasing interest in targeting this process to normalize the tumour microenvironment and thus enhance the treatment of solid tumours. Herein, we review advances made in unravelling the complexity of cancer-associated fibroinflammation that can inform the rational design of therapies targeting this.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Duan L, Cao S, Zhao F, Du X, Gao Z, Wang X, Bian F. Effects of FAP+ fibroblasts on cell proliferation migration and immunoregulation of esophageal squamous carcinoma cells through the CXCL12/CXCR4 axis. Mol Cell Biochem 2025; 480:3841-3855. [PMID: 39934460 DOI: 10.1007/s11010-025-05226-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Cancer-associated fibroblasts (CAFs) secrete and synthesize fibroblast activation protein (FAP), which could promote proliferation and immunosuppression of multiple cancers including esophageal squamous cell carcinoma (ESCC). CXCL12/CXCR4 signaling could be revitalized by CAFs in cancer cells. Nevertheless, the significance of this interaction in ESCC has yet to be elucidated. Herein, we investigated whether FAP+ CAF cells could promote ESCC cells proliferation, migration and regulate immunity through the CXCL12/CXCR4 pathway in vitro and in vivo. The protein expression level of FSP1, FAP, CD8+ and Ki-67 in different sample was estimated by IHC and western blot. qPCR was used to quantify the mRNA level of FSP1, FAP, CD8+ and Ki-67 in different sample. The cell viability, proliferation, migration and invasion of different sample were evaluated by CCK-8, EdU staining, wound healing assay and Transwell assay, respectively. The ELISA was carried out to measure the protein level of IFN-γ, TNF-α, GZMB and IL-2. ESCC xenograft mice model was established to assess the impact of FAP+ CAF. FSP1, FAP, CD8+ and Ki-67 are greatly up-regulated in hESCC tissues. Through CXCL12/CXCR4 axis, FAP-positive CAF was capable of promoting the cell proliferation, migration and invasion of ESCC tumor cells and preventing the CD8+ T cells from secreting cytokine. Blocking this signaling with selective CXCR4 antagonist could counteract the effects caused by high-expression of FAP. FAP+ CAFs could inhibit the occurrence and development of tumors. These results indicated that FAP-positive CAF have an impact on cell proliferation migration and immunoregulation of ESCC through the CXCL12/CXCR4 axis.
Collapse
Affiliation(s)
- Lijuan Duan
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China.
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China.
| | - Shasha Cao
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China
| | - Fang Zhao
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China
| | - Xianjuan Du
- Department of Pathology, Anyang Cancer Hospital, Anyang, 455000, Henan Province, People's Republic of China
| | - Zhaowei Gao
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China
| | - Xiaoxiao Wang
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
| | - Fang Bian
- Department of Pathology, Anyang Cancer Hospital, Anyang, 455000, Henan Province, People's Republic of China
| |
Collapse
|
3
|
Gacha-Garay MJ, Liu H, Evans SE, Mills TW, Chen J. Apoptosis inhibition reprograms alveolar myofibroblasts toward ductal myofibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.26.654588. [PMID: 40492191 PMCID: PMC12148055 DOI: 10.1101/2025.05.26.654588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
The epithelial tree of the lung is shaped proximo-distally by airway smooth muscle cells (ASMCs), ductal myofibroblasts (DMFs), and, transiently, alveolar myofibroblasts (AMFs). Lineage tracing and snapshot imaging suggest the clearance of AMFs via apoptosis post-alveologenesis, although definitive evidence is lacking. Here, we generate an inducible BCL2 overexpression mouse allele to inhibit AMF apoptosis. Using three independent Cre drivers and single-cell RNA-seq, we show that BCL2-rescued AMFs persist around distal alveolar ducts and alveoli and, unexpectedly, mature toward DMFs. Both normal DMFs and rescued DMF-like cells upregulate contractile proteins in a house dust mite-induced asthma model. Our findings demonstrate the apoptotic clearance of AMFs, as well as fate plasticity and pathophysiological convergence of lung mesenchymal cells of the epithelial axis.
Collapse
|
4
|
Li D, Rudloff U. Emerging therapeutics targeting tumor-associated macrophages for the treatment of solid organ cancers. Expert Opin Emerg Drugs 2025:1-39. [PMID: 40353504 DOI: 10.1080/14728214.2025.2504376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Over the last decade, immune checkpoint inhibitors (ICIs) like PD-1/PD-L1 or CTLA-4, which reinvigorate T cells for tumor control have become standard-of-care treatment options. In response to the increasingly recognized mechanisms of resistance to T cell activation in immunologically cold tumors, immuno-oncology drug development has started to shift beyond T cell approaches. These include tumor-associated macrophages (TAMs), a major pro-tumor immune cell population in the tumor microenvironment known to silence immune responses. AREAS COVERED Here we outline anti-TAM therapies in current development, either as monotherapy or in combination with other treatment modalities. We describe emerging drugs targeting TAMs under investigation in phase II and III testing with a focus on their distinguishing mechanism of action which include (1) reprogramming of TAMs toward anti-tumor function and immune surveillance, (2) blockade of recruitment, and (3) reduction and ablation of TAMs. EXPERT OPINION Several new immuno-oncology agents are under investigation to harness anti-tumor functions of TAMs. While robust anti-tumor efficacy of anti-TAM therapies across advanced solid organ cancers remains elusive to-date, TAM reprogramming therapies have yielded benefits in select cancers. The inherent heterogeneity of the diverse TAM population will require enhanced investments into biomarker-driven approaches to fully leverage its therapeutic potential.
Collapse
Affiliation(s)
- Dandan Li
- Developmental Therapeutics Branch (TDB), Biology Group, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Udo Rudloff
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
5
|
Gaspard-Boulinc LC, Gortana L, Walter T, Barillot E, Cavalli FMG. Cell-type deconvolution methods for spatial transcriptomics. Nat Rev Genet 2025:10.1038/s41576-025-00845-y. [PMID: 40369312 DOI: 10.1038/s41576-025-00845-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
Spatial transcriptomics is a powerful method for studying the spatial organization of cells, which is a critical feature in the development, function and evolution of multicellular life. However, sequencing-based spatial transcriptomics has not yet achieved cellular-level resolution, so advanced deconvolution methods are needed to infer cell-type contributions at each location in the data. Recent progress has led to diverse tools for cell-type deconvolution that are helping to describe tissue architectures in health and disease. In this Review, we describe the varied types of cell-type deconvolution methods for spatial transcriptomics, contrast their capabilities and summarize them in a web-based, interactive table to enable more efficient method selection.
Collapse
Affiliation(s)
- Lucie C Gaspard-Boulinc
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Luca Gortana
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Thomas Walter
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Emmanuel Barillot
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Florence M G Cavalli
- Institut Curie, PSL University, Paris, France.
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France.
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France.
| |
Collapse
|
6
|
Joo H, Olea XD, Zhuang A, Zheng B, Kim H, Ronai ZA. Epigenetic mechanisms in melanoma development and progression. Trends Cancer 2025:S2405-8033(25)00099-8. [PMID: 40328568 DOI: 10.1016/j.trecan.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025]
Abstract
Knowledge of cancer development and progression gained over the last few decades has enabled mapping of genetic and epigenetic changes unique to different phases of tumor evolution. Here we focus on epigenetic changes that drive melanoma development and progression. We highlight the importance of epigenetic mechanisms which encompass crosstalk with melanoma microenvironment that affect metastasis and therapy resistance. This review summarizes recent advances and describes potential strategies to leverage this knowledge to devise new therapies.
Collapse
Affiliation(s)
- Hyunjeong Joo
- Jim and Eleanor Randall Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ximena Diaz Olea
- Jim and Eleanor Randall Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Aojia Zhuang
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Bin Zheng
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Hyungsoo Kim
- Jim and Eleanor Randall Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ze'ev A Ronai
- Jim and Eleanor Randall Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Translational Research Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
7
|
Seban RD, Buvat I, Champion L, Bidard FC, Kieffer Y, Vincent-Salomon A, Peltier A, Mechta-Grigoriou F. Beyond FAP: ANTXR1 as a novel target for PET imaging and radio-ligand therapy in immuno-oncology? Eur J Nucl Med Mol Imaging 2025; 52:1948-1950. [PMID: 39907795 DOI: 10.1007/s00259-025-07126-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Affiliation(s)
- Romain-David Seban
- Department of Nuclear Medicine, Institute of Women's Cancer, Institut Curie, 92210, Saint-Cloud, France.
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm U1288, PSL University, Paris Saclay University, Institute of Women's Cancer, Institut Curie, 91400, Orsay, France.
| | - Irene Buvat
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm U1288, PSL University, Paris Saclay University, Institute of Women's Cancer, Institut Curie, 91400, Orsay, France
| | - Laurence Champion
- Department of Nuclear Medicine, Institute of Women's Cancer, Institut Curie, 92210, Saint-Cloud, France
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm U1288, PSL University, Paris Saclay University, Institute of Women's Cancer, Institut Curie, 91400, Orsay, France
| | - Francois-Clement Bidard
- Department of Medical Oncology, Institute of Women's Cancer, Institut Curie, 75006, Paris, France
- Circulating Tumor Biomarkers Laboratory, SiRIC, PSL Research University, Institute of Women's Cancer, Institut Curie, Paris, France
| | - Yann Kieffer
- Stress and Cancer Laboratory, Equipe Labélisée Par La Ligue Nationale Contre Le Cancer, Inserm U1339 - UMR3666 CNRS, PSL Research University, Institute of Women's Cancer, Institut Curie, 26, rue d'Ulm, F-75248, Paris, France
| | - Anne Vincent-Salomon
- Department of Diagnostic and Theranostic Medicine, Institute of Women's Cancer, Institut Curie, 75006, Paris, France
| | - Agathe Peltier
- Stress and Cancer Laboratory, Equipe Labélisée Par La Ligue Nationale Contre Le Cancer, Inserm U1339 - UMR3666 CNRS, PSL Research University, Institute of Women's Cancer, Institut Curie, 26, rue d'Ulm, F-75248, Paris, France
| | - Fatima Mechta-Grigoriou
- Stress and Cancer Laboratory, Equipe Labélisée Par La Ligue Nationale Contre Le Cancer, Inserm U1339 - UMR3666 CNRS, PSL Research University, Institute of Women's Cancer, Institut Curie, 26, rue d'Ulm, F-75248, Paris, France
| |
Collapse
|
8
|
Chen W, Jiang M, Zou X, Chen Z, Shen L, Hu J, Kong M, Huang J, Ni C, Xia W. Fibroblast Activation Protein (FAP) + cancer-associated fibroblasts induce macrophage M2-like polarization via the Fibronectin 1-Integrin α5β1 axis in breast cancer. Oncogene 2025:10.1038/s41388-025-03359-3. [PMID: 40263422 DOI: 10.1038/s41388-025-03359-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 04/24/2025]
Abstract
Cancer-associated fibroblasts expressing fibroblast activation protein (FAP+ CAFs) are critical modulators of the breast cancer microenvironment, yet their immunoregulatory mechanisms remain poorly understood. Through integrated analysis of single-cell RNA sequencing data, clinical specimens, and in vivo and in vitro experiments, we identified FAP+ CAFs as the predominant stromal population associated with poor clinical outcomes and immunosuppressive features. Mechanistically, FAP+ CAFs secrete high levels of fibronectin 1 (FN1), which engages integrin α5β1 on macrophages to trigger FAK-AKT-STAT3 signaling, driving their polarization toward an immunosuppressive M2-like phenotype. Importantly, pharmacological disruption of FN1-integrin α5β1 signaling using Cilengitide effectively reprogrammed the tumor immune landscape and suppressed tumor growth in mice models. These findings establish FAP+ CAF-derived FN1 as a critical orchestrator of tumor immunosuppression and identify the FN1-integrin α5β1 axis as a promising therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Wuzhen Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Oncology, Lanxi People's Hospital, Jinhua, Zhejiang, China
| | - Mengjie Jiang
- Department of Radiotherapy, First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Xinbo Zou
- Department of Otolaryngology, First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Zhigang Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lesang Shen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianming Hu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mingxiang Kong
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Jian Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| | - Chao Ni
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| | - Wenjie Xia
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Liu Z, Ba Y, Shan D, Zhou X, Zuo A, Zhang Y, Xu H, Liu S, Liu B, Zhao Y, Weng S, Wang R, Deng J, Luo P, Cheng Q, Hu X, Yang S, Wang F, Han X. THBS2-producing matrix CAFs promote colorectal cancer progression and link to poor prognosis via the CD47-MAPK axis. Cell Rep 2025; 44:115555. [PMID: 40222008 DOI: 10.1016/j.celrep.2025.115555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/30/2024] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) display significant functional and molecular heterogeneity within the tumor microenvironment, playing diverse roles in cancer progression. Employing single-cell RNA sequencing data of colorectal cancer (CRC), we identified a subset of matrix CAFs (mCAFs) as a critical subtype that secretes THBS2, a molecule linked to advanced cancer stages and poor prognosis. Spatial transcriptomics and multiplex immunohistochemistry revealed clear spatial colocalization between THBS2-producing mCAFs and tumor cells. Mechanically, CAF-secreted THBS2 binds to CD47 on tumor cells, triggering the MAPK/ERK5 signaling pathway, which enhances tumor progression. The tumor-promoting role of THBS2 was further validated using fibroblast-specific THBS2 knockout mice, patient-derived organoids, and xenografts. Moreover, the transcription factor CREB3L1 was identified as a regulator of the transformation of normal fibroblasts into THBS2-producing mCAFs. These findings underscore the pivotal role of THBS2 in CRC progression and highlight the therapeutic potential of targeting the THBS2-CD47 axis and CREB3L1 in CRC.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Dan Shan
- Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YT, UK
| | - Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Benyu Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanan Zhao
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ruizhi Wang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jinhai Deng
- Richard Dimbleby Department of Cancer Research, Comprehensive Cancer Centre, Kings College London, London, UK
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Hu
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China; Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, China.
| | - Fubing Wang
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China; Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China; Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China.
| |
Collapse
|
10
|
Wang TC, Huang W, Li SX, Li JX, Zhou P, Wang LZ, Wei N, Cai WW, Hu JJ, Xiao YD. High Fibroblast Activation Protein Expression in Hepatocellular Carcinoma: CT Imaging Features and Histological Characteristics. Acad Radiol 2025:S1076-6332(25)00315-0. [PMID: 40254479 DOI: 10.1016/j.acra.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: 03/04/2025] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
RATIONALE AND OBJECTIVES To clarify computed tomography (CT) imaging features of high fibroblast activation protein (FAP) expression in hepatocellular carcinoma (HCC) and to investigate their correlation to histological characteristics and prognosis. METHODS This retrospective multicenter study evaluated patients with HCC who underwent liver resection between August 2013 and June 2023. Histological staining for FAP was performed, and patients were classified into low and high-FAP expression groups. A predictive model was developed and validated to identify FAP expression levels among CT imaging features. Moreover, CT imaging-related histological characteristics were evaluated. With the predictive model, patients in training cohort were stratified into predicted low and high-FAP expression groups. Overall survival (OS) and recurrence-free survival (RFS) were compared accordingly. Besides, propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) analysis were performed, and OS and RFS between the two groups were also compared. RESULTS In total, 1197 patients were included (high-FAP HCCs, n=267). Delayed central enhancement (OR: 15.196, 95% CI: 8.996-25.670, P<0.001) and dilated vasculature (OR: 7.455, 95% CI: 4.928-11.277, P<0.001) independently predicted high-FAP expression (AUCs: 0.779, training; 0.766, external validation). Based on these two CT imaging features, intratumoral fibrosis (ITF) and vessel-encapsulating tumor cluster (VETC) pattern were determined. Compared to low-FAP HCCs, high-FAP HCCs were more frequently with high ITF grade (54.7% vs. 22.4%; P<0.001) and VETC pattern (52.1% vs. 22.6%; P<0.001). Patients were divided into predicted low (n=564) and high (n=262) FAP expression groups. The predicted low-FAP group had significantly better or favorable trend toward improved OS and RFS than predicted high-FAP group before (P<0.001, OS and RFS), and after PSM (P=0.009, OS; P=0.005, RFS) and IPTW (P=0.019, OS; P=0.077, RFS). CONCLUSION Delayed central enhancement and dilated vasculature are independent factors of high-FAP expression in HCC. Noninvasive identifying FAP expression may offer potential prognostic and therapeutic insights.
Collapse
Affiliation(s)
- Tian-Cheng Wang
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Wei Huang
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Shu-Xian Li
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Jun-Xiang Li
- Department of Interventional Radiology, Guizhou Medical University Affiliated Cancer Hospital, Guiyang 550004, China (J.X.L.)
| | - Peng Zhou
- Department of Pathology, the Second Xiangya Hospital of Central South University, Changsha 410011, China (P.Z.)
| | - Li-Zhou Wang
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China (L.Z.W.)
| | - Nan Wei
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg 69120, Germany (N.W.)
| | - Wen-Wu Cai
- Department of Liver Surgery, the Second Xiangya Hospital of Central South University, Changsha 410011, China (W.W.C.)
| | - Jun-Jiao Hu
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Yu-Dong Xiao
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.).
| |
Collapse
|
11
|
Kay EJ, Zanivan S. The tumor microenvironment is an ecosystem sustained by metabolic interactions. Cell Rep 2025; 44:115432. [PMID: 40088447 DOI: 10.1016/j.celrep.2025.115432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/09/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) and immune cells make up two major components of the tumor microenvironment (TME), contributing to an ecosystem that can either support or restrain cancer progression. Metabolism is a key regulator of the TME, providing a means for cells to communicate with and influence each other, modulating tumor progression and anti-tumor immunity. Cells of the TME can metabolically interact directly through metabolite secretion and consumption or by influencing other aspects of the TME that, in turn, stimulate metabolic rewiring in target cells. Recent advances in understanding the subtypes and plasticity of cells in the TME both open up new avenues and create challenges for metabolically targeting the TME to hamper tumor growth and improve response to therapy. This perspective explores ways in which the CAF and immune components of the TME could metabolically influence each other, based on current knowledge of their metabolic states, interactions, and subpopulations.
Collapse
Affiliation(s)
- Emily Jane Kay
- Cancer Research UK Scotland Institute, Glasgow G61 1BD, UK.
| | - Sara Zanivan
- Cancer Research UK Scotland Institute, Glasgow G61 1BD, UK; School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK; Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Li F, Gao C, Huang Y, Qiao Y, Xu H, Liu S, Wu H. Unraveling the breast cancer tumor microenvironment: crucial factors influencing natural killer cell function and therapeutic strategies. Int J Biol Sci 2025; 21:2606-2628. [PMID: 40303301 PMCID: PMC12035885 DOI: 10.7150/ijbs.108803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/04/2025] [Indexed: 05/02/2025] Open
Abstract
Natural killer (NK) cells have emerged as a novel and effective treatment for breast cancer. Nevertheless, the breast cancer tumor microenvironment (TME) manifests multiple immunosuppressive mechanisms, impeding the proper execution of NK cell functions. This review summarizes recent research on the influence of the TME on the functionality of NK cells in breast cancer. It delves into the effects of the internal environment of the TME on NK cells and elucidates the roles of diverse stromal components, immune cells, and signaling molecules in regulating NK cell activity within the TME. It also summarizes therapeutic strategies based on small-molecule inhibitors, antibody therapies, and natural products, as well as the progress of research in preclinical and clinical trials. By enhancing our understanding of the immunosuppressive TME and formulating strategies to counteract its effects, we could fully harness the therapeutic promise of NK cells in breast cancer treatment.
Collapse
Affiliation(s)
- Feifei Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunfang Gao
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Huang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| | - Yu Qiao
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| | - Hongxiao Xu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Liu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huangan Wu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| |
Collapse
|
13
|
Song J, Wei R, Liu C, Zhao Z, Liu X, Wang Y, Liu F, Liu X. Antigen-presenting cancer associated fibroblasts enhance antitumor immunity and predict immunotherapy response. Nat Commun 2025; 16:2175. [PMID: 40038297 PMCID: PMC11880398 DOI: 10.1038/s41467-025-57465-7] [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: 12/07/2023] [Accepted: 02/23/2025] [Indexed: 03/06/2025] Open
Abstract
Cancer-associated fibroblasts (CAF) play a crucial role in tumor progression and immune regulation. However, the functional heterogeneity of CAFs remains unclear. Here, we identify antigen-presenting CAFs (apCAF), characterized by high MHC II expression, in gastric cancer (GC) tumors and find that apCAFs are preferentially located near tertiary lymphoid structures. Both in vivo and in vitro experiments demonstrate that apCAFs promote T cell activation and enhances its cytotoxic and proliferative capacities, thereby strengthening T cell-mediated anti-tumor immunity. Additionally, apCAFs facilitate the polarization of macrophages toward a pro-inflammatory phenotype. These polarized macrophages, in turn, promote the formation of apCAFs, creating a positive feedback loop that amplifies anti-tumor immune responses. Notably, baseline tumors in immunotherapy responders across various cancer types exhibit higher levels of apCAFs infiltration. This study advances the understanding of CAFs heterogeneity in GC and highlights apCAFs as a potential biomarker for predicting immunotherapy response in pan-cancer.
Collapse
Affiliation(s)
- Junquan Song
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Rongyuan Wei
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Chenchen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Zhenxiong Zhao
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xuanjun Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yanong Wang
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
| | - Fenglin Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
| | - Xiaowen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Zhong B, Du J, Liu F, Sun S. The Role of Yes-Associated Protein in Inflammatory Diseases and Cancer. MedComm (Beijing) 2025; 6:e70128. [PMID: 40066231 PMCID: PMC11892025 DOI: 10.1002/mco2.70128] [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: 04/17/2024] [Revised: 02/02/2025] [Accepted: 02/11/2025] [Indexed: 03/17/2025] Open
Abstract
Yes-associated protein (YAP) plays a central role in the Hippo pathway, primarily governing cell proliferation, differentiation, and apoptosis. Its significance extends to tumorigenesis and inflammatory conditions, impacting disease initiation and progression. Given the increasing relevance of YAP in inflammatory disorders and cancer, this study aims to elucidate its pathological regulatory functions in these contexts. Specifically, we aim to investigate the involvement and molecular mechanisms of YAP in various inflammatory diseases and cancers. We particularly focus on how YAP activation, whether through Hippo-dependent or independent pathways, triggers the release of inflammation and inflammatory mediators in respiratory, cardiovascular, and digestive inflammatory conditions. In cancer, YAP not only promotes tumor cell proliferation and differentiation but also modulates the tumor immune microenvironment, thereby fostering tumor metastasis and progression. Additionally, we provide an overview of current YAP-targeted therapies. By emphasizing YAP's role in inflammatory diseases and cancer, this study aims to enhance our understanding of the protein's pivotal involvement in disease processes, elucidate the intricate pathological mechanisms of related diseases, and contribute to future drug development strategies targeting YAP.
Collapse
Affiliation(s)
- Bing Zhong
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jintao Du
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Feng Liu
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Silu Sun
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| |
Collapse
|
15
|
Chang ACC, Schlegel BT, Carleton N, McAuliffe PF, Oesterreich S, Schwartz R, Lee AV. CITEgeist: Cellular Indexing of Transcriptomes and Epitopes for Guided Exploration of Intrinsic Spatial Trends. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.15.638331. [PMID: 40027773 PMCID: PMC11870549 DOI: 10.1101/2025.02.15.638331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Spatial transcriptomics provides insights into tissue architecture by linking gene expression with spatial localization. Current deconvolution methods rely heavily on single-cell RNA sequencing (scRNA-seq) references, which are costly and often unavailable, mainly if the tissue under evaluation is limited, such as in a core biopsy specimen. We present a novel tool, CITEgeist, that deconvolutes spatial transcriptomics data using antibody capture from the same slide as the reference, directly leveraging cell surface protein measurements from the same tissue section. This approach circumvents the limitations of scRNA-seq as a reference, offering a cost-effective and biologically grounded alternative. Our method employs mathematical optimization to estimate cell type proportions and gene expression profiles, incorporating sparsity constraints for robustness and interpretability. Benchmarks against state-of-the-art deconvolution methods show improved accuracy in cell type resolution, particularly in dense tumor microenvironments, while maintaining computational efficiency. This antibody-based tool advances spatial transcriptomics by providing a scalable, accurate, and reference-independent solution for deconvolution in complex tissues. We validate this tool by using a combined approach of simulated data and clinical samples by applying CITEgeist to translational pre-treatment and post-treatment ER+ breast tumors from an ongoing clinical trial, emphasizing the applicability and robustness of CITEgeist.
Collapse
Affiliation(s)
- Alexander Chih-Chieh Chang
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Magee-Womens Research Institute, Pittsburgh PA, USA
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Brent T. Schlegel
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Magee-Womens Research Institute, Pittsburgh PA, USA
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neil Carleton
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Magee-Womens Research Institute, Pittsburgh PA, USA
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Priscilla F. McAuliffe
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Magee-Womens Research Institute, Pittsburgh PA, USA
- Department of Surgery, Division of Breast Surgical Oncology, University of Pittsburgh School of Medicine, Pittsburgh PA, USA
| | - Steffi Oesterreich
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Magee-Womens Research Institute, Pittsburgh PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
| | - Russell Schwartz
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Adrian V. Lee
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Magee-Womens Research Institute, Pittsburgh PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
- Institute of Precision Medicine, Pittsburgh PA, USA
| |
Collapse
|
16
|
Horenstein AL, Faini AC, Morandi F, Ortolan E, Storti P, Giuliani N, Richardson PG, Malavasi F. Monoclonal anti-CD38 therapy in human myeloma: retrospects and prospects. Front Immunol 2025; 16:1519300. [PMID: 40013150 PMCID: PMC11860881 DOI: 10.3389/fimmu.2025.1519300] [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: 10/29/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025] Open
Abstract
Monoclonal antibody therapy using CD38 as a target remains central to managing human multiple myeloma (MM). CD38 was selected early on as a target for mAb-mediated therapy for MM, driven by findings from an early Cluster of Differentiation (CD) Workshop. The first CD38-targeting antibody to be approved yielded strong trial results, significantly improving survival rates and earning widespread patient acceptance. However, resistance to the therapy later emerged, complicating treatment management. Despite CD38's still central role in MM therapy, too little attention has been paid to its broader roles-not only as a myeloma marker but also as an enzyme and adhesion molecule in physiology. This review, a collaborative effort between basic scientists and clinical experts, explores some of the lesser-known mechanisms of antibody action and interactions with CD38 at key stages of treatment. The review also highlights the relevance of the MM environment, focusing on the importance of the bone marrow (BM) niche. The goal is to identify new agents whose unique properties may enhance tumor eradication. By gaining a deeper understanding of interactions between therapeutic antibodies, myeloma cells, and the tumor microenvironment (TME), it is hoped that previously unrecognized vulnerabilities within the disease may be revealed, paving the way to more effective treatment strategies.
Collapse
Affiliation(s)
- Alberto L. Horenstein
- Lab of Immunogenetics, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Angelo C. Faini
- Lab of Immunogenetics, Department of Medical Sciences, University of Torino, Torino, Italy
- Immunogenetics and Transplant Biology, University Hospital “Città della Salute e della Scienza di Torino”, Torino, Italy
| | - Fabio Morandi
- UOSD Laboratorio di Terapie Cellulari, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Erika Ortolan
- Lab of Immunogenetics, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Paola Storti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma & Multiple Myeloma Program, AOU, Parma, Italy
| | - Paul G. Richardson
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Fabio Malavasi
- Lab of Immunogenetics, Department of Medical Sciences, University of Torino, Torino, Italy
- Fondazione Ricerca Molinette Ets, Torino, Italy
| |
Collapse
|
17
|
Garate-Soraluze E, Serrano-Mendioroz I, Fernández-Rubio L, De Andrea CE, Barrio-Alonso C, Herrero CDP, Teijeira A, Luri-Rey C, Claus C, Tanos T, Klein C, Umana P, Rullan A, Simón JA, Collantes M, Sánchez-Mateos P, Melero I, Rodriguez-Ruiz ME. 4-1BB agonist targeted to fibroblast activation protein α synergizes with radiotherapy to treat murine breast tumor models. J Immunother Cancer 2025; 13:e009852. [PMID: 39933836 PMCID: PMC11815443 DOI: 10.1136/jitc-2024-009852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 11/01/2024] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Ionizing radiation (IR) is a double-edged sword for immunotherapy as it may have both immunosuppressive and immunostimulatory effects. The biological effects of IR on the tumor microenvironment (TME) are a key factor for this balance. Fibroblast activation protein (FAP) is expressed on the surface of cancer-associated fibroblasts (CAF) in many cancer types and its abundance is associated with the poor immune response to immune-checkpoint-blockade in patients. We hypothesized that IR increases FAP expression in CAFs, therefore the combination of IR with targeted immunomodulators such as an agonistic anti-FAP-4-1BBL fusion protein could enhance the immune-mediated antitumoral effects of these treatments. METHODS The murine transplantable TS/A tumor-cell-line co-engrafted with CAFs was used to investigate increases in FAP expression in tumors following irradiation using immunohistochemistry, real-time polymerase chain reaction (RT-PCR) and multiplex tissue immunofluorescence. One lesion of bilateral tumor-bearing mice was only locally irradiated or combined with weekly injections of the bispecific muFAP-4-1BBL fusion protein (a mouse surrogate for RG7826). Tumor sizes were followed over time and TME was assessed by flow cytometry. Selective monoclonal antibody (mAb)-mediated depletions of immune cell populations, neutralizing interferon alpha/beta receptor 1 (IFNAR-I) IFNAR and interferon (IFN)-γ mAbs and gene-modified mice (4-1BB-/-) were used to delineate the immune cell subsets and mechanisms required for efficacy. 67Ga labeled muFAP-4-1BBL tracked by SPECT-CT was used to study biodistribution. In human colorectal carcinoma samples, the inducibility of FAP expression following radiotherapy was explored by multiplex immunofluorescence. RESULTS Irradiation of TS/A+CAF tumors in mice showed an increase in FAP levels after local irradiation. A suboptimal radiotherapy regimen in combination with muFAP-4-1BBL attained primary tumor control and measurable abscopal effects. Immune TME landscape analyses showed post-treatment increased infiltration of activated immune cells associated with the combined radioimmunotherapy treatment. Efficacy depended on CD8+ T cells, type I IFN, IFN-γ and ability to express 4-1BB. Biodistribution studies of muFAP-4-1BBL indicated enriched tumor targeting to irradiated tumors. Human colorectal cancer samples pre and post irradiation showed enhanced FAP expression after radiotherapy. CONCLUSION Increased FAP expression in the TME as a result of radiotherapy can be exploited to target agonist 4-1BB immunotherapy to malignant tumor lesions using an FAP-4-1BBL antibody fusion protein.
Collapse
Affiliation(s)
- Eneko Garate-Soraluze
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Irantzu Serrano-Mendioroz
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Leticia Fernández-Rubio
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Carlos E De Andrea
- Department of Pathology, Cancer Center Clinica Universidad de Navarra, Pamplona, Spain
| | | | | | - Alvaro Teijeira
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Carlos Luri-Rey
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Christina Claus
- Roche Innovation Centre Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Tamara Tanos
- Roche Innovation Centre Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Centre Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Pablo Umana
- Roche Innovation Centre Zurich, Roche Pharma Research and Early Development (pRED), Schlieren, Switzerland
| | - Antonio Rullan
- Institute of Immunology and Transplantation, University College London, London, UK
| | - Jon Ander Simón
- Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| | - María Collantes
- Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| | | | - Ignacio Melero
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Departments of Immunology-Immunotherapy and Radiation Oncology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Institute for Health Research (IDISNA), Pamplona, Spain
| | - Maria E Rodriguez-Ruiz
- Department of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Departments of Immunology-Immunotherapy and Radiation Oncology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Institute for Health Research (IDISNA), Pamplona, Spain
| |
Collapse
|
18
|
Corvigno S, Fernebro J, Karlsson JS, Mezheieusky A, Martín-Bernabé A, De La Fuente LM, Westbom-Fremer S, Carlson JW, Klein C, Kannisto P, Hedenfalk I, Malander S, Östman A, Dahlstrand H. High prevalence of FAP+ cancer-associated fibroblasts predicts poor outcome in patients with high-grade serous ovarian cancer with high CD8 T-cell density. Gynecol Oncol 2025; 193:148-155. [PMID: 39914230 DOI: 10.1016/j.ygyno.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 03/03/2025]
Abstract
OBJECTIVE Studies have implied that fibroblasts may act as regulators of immune cells in the tumor microenvironment (TME). We investigated the clinical relevance of fibroblast activation protein (FAP) positive stroma in high-grade serous ovarian cancer (HGSC) in relation to CD8+ lymphocyte's infiltration. METHODS In a discovery cohort (N = 113) of HGSC, expression of FAP and CD8 in the TME was analyzed with immunohistochemistry. Results were correlated with overall survival (OS) and progression-free survival (PFS). The findings were validated in an independent cohort of HGSC (N = 121) and in public available datasets. RESULTS High infiltration of CD8+ cells in the TME of HGSC was found to be associated with longer OS, as previously known. Increased expression of FAP was associated with shorter median PFS (11.4 vs. 18.6 months) in tumors with high density of CD8+ cells (HR 4.03, CI 95 % 1.38-11.72, p = 0.01). Similarly, in the validation cohort, high intensity of FAP in cases with high density of CD8+ cells was associated with shorter OS, 31.5 vs 76.9 months (HR 2.83; 95 % CI 1.17-6.86, p = 0.02). The results were consistent in multivariable analyses. The association between high FAP expression and poor outcome in high density CD8 HGSC was also confirmed in publicly available datasets. CONCLUSIONS The TME infiltration of FAP-positive fibroblasts is associated with poor prognosis in HGSC with high CD8+ cells density. Targeting the FAP+ subset of fibroblasts may unlock the local immune-activation in the TME thus enhance the positive prognostic effect of T-cells in ovarian cancer.
Collapse
Affiliation(s)
- Sara Corvigno
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Josefin Fernebro
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Gynecologic Oncology, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Josefin Severin Karlsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden.
| | - Artur Mezheieusky
- IGP, Uppsala University, Sweden; Vall d'Hebron Institute of Oncology, Molecular oncology group, Barcelona, Spain
| | | | - Laura Martin De La Fuente
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Sofia Westbom-Fremer
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Joseph W Carlson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Pathology and Laboratory Medicine, University of Southern California, Los Angeles, USA
| | | | - Paivi Kannisto
- Department of Obstetrics and Gynecology, Skåne University Hospital and Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences Lund, Lund University and University Hospital, Lund, Sweden
| | - Susanne Malander
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Hanna Dahlstrand
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Gynecologic Oncology, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
19
|
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
|
20
|
Hanahan D, Michielin O, Pittet MJ. Convergent inducers and effectors of T cell paralysis in the tumour microenvironment. Nat Rev Cancer 2025; 25:41-58. [PMID: 39448877 DOI: 10.1038/s41568-024-00761-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
Tumorigenesis embodies the formation of a heterotypic tumour microenvironment (TME) that, among its many functions, enables the evasion of T cell-mediated immune responses. Remarkably, most TME cell types, including cancer cells, fibroblasts, myeloid cells, vascular endothelial cells and pericytes, can be stimulated to deploy immunoregulatory programmes. These programmes involve regulatory inducers (signals-in) and functional effectors (signals-out) that impair CD8+ and CD4+ T cell activity through cytokines, growth factors, immune checkpoints and metabolites. Some signals target specific cell types, whereas others, such as transforming growth factor-β (TGFβ) and prostaglandin E2 (PGE2), exert broad, pleiotropic effects; as signals-in, they trigger immunosuppressive programmes in most TME cell types, and as signals-out, they directly inhibit T cells and also modulate other cells to reinforce immunosuppression. This functional diversity and redundancy pose a challenge for therapeutic targeting of the immune-evasive TME. Fundamentally, the commonality of regulatory programmes aimed at abrogating T cell activity, along with paracrine signalling between cells of the TME, suggests that many normal cell types are hard-wired with latent functions that can be triggered to prevent inappropriate immune attack. This intrinsic capability is evidently co-opted throughout the TME, enabling tumours to evade immune destruction.
Collapse
Affiliation(s)
- Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
| | - Olivier Michielin
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
| | - Mikael J Pittet
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva (UNIGE), Geneva, Switzerland
| |
Collapse
|
21
|
An J, Lu Y, Chen Y, Chen Y, Zhou Z, Chen J, Peng C, Huang R, Peng F. Spatial transcriptomics in breast cancer: providing insight into tumor heterogeneity and promoting individualized therapy. Front Immunol 2024; 15:1499301. [PMID: 39749323 PMCID: PMC11693744 DOI: 10.3389/fimmu.2024.1499301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
A comprehensive understanding of tumor heterogeneity, tumor microenvironment and the mechanisms of drug resistance is fundamental to advancing breast cancer research. While single-cell RNA sequencing has resolved the issue of "temporal dynamic expression" of genes at the single-cell level, the lack of spatial information still prevents us from gaining a comprehensive understanding of breast cancer. The introduction and application of spatial transcriptomics addresses this limitation. As the annual technical method of 2020, spatial transcriptomics preserves the spatial location of tissues and resolves RNA-seq data to help localize and differentiate the active expression of functional genes within a specific tissue region, enabling the study of spatial location attributes of gene locations and cellular tissue environments. In the context of breast cancer, spatial transcriptomics can assist in the identification of novel breast cancer subtypes and spatially discriminative features that show promise for individualized precise treatment. This article summarized the key technical approaches, recent advances in spatial transcriptomics and its applications in breast cancer, and discusses the limitations of current spatial transcriptomics methods and the prospects for future development, with a view to advancing the application of this technology in clinical practice.
Collapse
Affiliation(s)
- Junsha An
- West China School of Pharmacy, Sichuan University, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Cardiovascular Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yajie Lu
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yuxi Chen
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yuling Chen
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhaokai Zhou
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ruizhen Huang
- Cardiovascular Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
de la Jara Ortiz F, Cimmino C, Ventre M, Cambi A. Understanding and measuring mechanical signals in the tumor stroma. FEBS Open Bio 2024. [PMID: 39523476 DOI: 10.1002/2211-5463.13923] [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: 06/30/2024] [Revised: 09/30/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
The tumor microenvironment (TME) is well known for its immune suppressive role, especially in solid tumors which are characterized by a thick, dense stroma. Apart from cell-cell interactions and biochemical signals, the tumor stroma is also characterized by its distinct mechanical properties, which are dictated by the composition and architecture of its extracellular matrix (ECM). Cancer-associated fibroblasts (CAFs) are the main producers and remodelers of the stromal ECM, and their heterogeneity has recently become a focus of intense research. This review describes recent findings highlighting CAF subtypes and their specific functions, as well as the development of 3D models to study tumor stroma mechanics in vitro. Finally, we discuss the quantitative techniques used to measure tissue mechanical properties at different scales. Given the diagnostic and prognostic value of stroma stiffness and composition, and the recent development of anti-tumor therapeutic strategies targeting the stroma, understanding and measuring tumor stroma mechanical properties has never been more timely or relevant.
Collapse
Affiliation(s)
- Fàtima de la Jara Ortiz
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Chiara Cimmino
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Fondazione Istituto Italiano di Tecnologia, Naples, Italy
| | - Maurizio Ventre
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Fondazione Istituto Italiano di Tecnologia, Naples, Italy
- Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy
| | - Alessandra Cambi
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
23
|
Zhang Y, Gong S, Liu X. Spatial transcriptomics: a new frontier in accurate localization of breast cancer diagnosis and treatment. Front Immunol 2024; 15:1483595. [PMID: 39439806 PMCID: PMC11493667 DOI: 10.3389/fimmu.2024.1483595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Breast cancer is one of the most prevalent cancers in women globally. Its treatment and prognosis are significantly influenced by the tumor microenvironment and tumor heterogeneity. Precision therapy enhances treatment efficacy, reduces unwanted side effects, and maximizes patients' survival duration while improving their quality of life. Spatial transcriptomics is of significant importance for the precise treatment of breast cancer, playing a critical role in revealing the internal structural differences of tumors and the composition of the tumor microenvironment. It offers a novel perspective in studying the spatial structure and cell interactions within tumors, facilitating more effective personalized treatments for breast cancer. This article will summarize the latest findings in the diagnosis and treatment of breast cancer from the perspective of spatial transcriptomics, focusing on the revelation of the tumor microenvironment, identification of new therapeutic targets, enhancement of disease diagnostic accuracy, comprehension of tumor progression and metastasis, assessment of drug responses, creation of high-resolution maps of tumor cells, representation of tumor heterogeneity, and support for clinical decision-making, particularly in elucidating the tumor microenvironment, tumor heterogeneity, immunotherapy and their correlation with clinical outcomes.
Collapse
Affiliation(s)
- Yang Zhang
- Breast and Thyroid Surgery, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Shuhua Gong
- Department of Student Affair, Shandong College of Traditional Chinese Medicine, Yantai, China
| | - Xiaofei Liu
- Breast and Thyroid Surgery, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| |
Collapse
|
24
|
Haberkorn U, Altmann A, Giesel FL, Kratochwil C. 1,090 Publications and 5 Years Later: Is FAP-Targeted Theranostics Really Happening? J Nucl Med 2024; 65:1518-1520. [PMID: 39168520 DOI: 10.2967/jnumed.124.267923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Affiliation(s)
- Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany;
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany
- Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany
| | - Annette Altmann
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany
| | - Frederik L Giesel
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Duesseldorf, Germany; and
- Institute for Radiation Sciences, Osaka University, Toyonaka, Japan
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
25
|
Xiong X, Wang X, Liu CC, Shao ZM, Yu KD. Deciphering breast cancer dynamics: insights from single-cell and spatial profiling in the multi-omics era. Biomark Res 2024; 12:107. [PMID: 39294728 PMCID: PMC11411917 DOI: 10.1186/s40364-024-00654-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/10/2024] [Indexed: 09/21/2024] Open
Abstract
As one of the most common tumors in women, the pathogenesis and tumor heterogeneity of breast cancer have long been the focal point of research, with the emergence of tumor metastasis and drug resistance posing persistent clinical challenges. The emergence of single-cell sequencing (SCS) technology has introduced novel approaches for gaining comprehensive insights into the biological behavior of malignant tumors. SCS is a high-throughput technology that has rapidly developed in the past decade, providing high-throughput molecular insights at the individual cell level. Furthermore, the advent of multitemporal point sampling and spatial omics also greatly enhances our understanding of cellular dynamics at both temporal and spatial levels. The paper provides a comprehensive overview of the historical development of SCS, and highlights the most recent advancements in utilizing SCS and spatial omics for breast cancer research. The findings from these studies will serve as valuable references for future advancements in basic research, clinical diagnosis, and treatment of breast cancer.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xin Wang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
26
|
Cords L, de Souza N, Bodenmiller B. Classifying cancer-associated fibroblasts-The good, the bad, and the target. Cancer Cell 2024; 42:1480-1485. [PMID: 39255773 DOI: 10.1016/j.ccell.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/16/2024] [Accepted: 08/09/2024] [Indexed: 09/12/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are heterogeneous and ubiquitous stromal cells within the tumor microenvironment (TME). Numerous CAF types have been described, typically using single-cell technologies such as single-cell RNA sequencing. There is no general classification system for CAFs, hampering their study and therapeutic targeting. We propose a simple CAF classification system based on single-cell phenotypes and spatial locations of CAFs in multiple cancer types, assess how our scheme fits within current knowledge, and invite the CAF research community to further refine it.
Collapse
Affiliation(s)
- Lena Cords
- University of Zurich, Department of Quantitative Biomedicine, Zurich, Switzerland; ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland
| | - Natalie de Souza
- University of Zurich, Department of Quantitative Biomedicine, Zurich, Switzerland; ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland; ETH Zurich, Institute of Molecular Systems Biology, Zurich, Switzerland
| | - Bernd Bodenmiller
- University of Zurich, Department of Quantitative Biomedicine, Zurich, Switzerland; ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland.
| |
Collapse
|
27
|
Xiang L, Rao J, Yuan J, Xie T, Yan H. Single-Cell RNA-Sequencing: Opening New Horizons for Breast Cancer Research. Int J Mol Sci 2024; 25:9482. [PMID: 39273429 PMCID: PMC11395021 DOI: 10.3390/ijms25179482] [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: 07/31/2024] [Revised: 08/25/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Breast cancer is the most prevalent malignant tumor among women with high heterogeneity. Traditional techniques frequently struggle to comprehensively capture the intricacy and variety of cellular states and interactions within breast cancer. As global precision medicine rapidly advances, single-cell RNA sequencing (scRNA-seq) has become a highly effective technique, revolutionizing breast cancer research by offering unprecedented insights into the cellular heterogeneity and complexity of breast cancer. This cutting-edge technology facilitates the analysis of gene expression profiles at the single-cell level, uncovering diverse cell types and states within the tumor microenvironment. By dissecting the cellular composition and transcriptional signatures of breast cancer cells, scRNA-seq provides new perspectives for understanding the mechanisms behind tumor therapy, drug resistance and metastasis in breast cancer. In this review, we summarized the working principle and workflow of scRNA-seq and emphasized the major applications and discoveries of scRNA-seq in breast cancer research, highlighting its impact on our comprehension of breast cancer biology and its potential for guiding personalized treatment strategies.
Collapse
Affiliation(s)
- Lingyan Xiang
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jie Rao
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ting Xie
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Honglin Yan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|