1
|
Ramos C, Gerakopoulos V, Oehler R. Metastasis-associated fibroblasts in peritoneal surface malignancies. Br J Cancer 2024; 131:407-419. [PMID: 38783165 PMCID: PMC11300623 DOI: 10.1038/s41416-024-02717-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Over decades, peritoneal surface malignancies (PSMs) have been associated with limited treatment options and poor prognosis. However, advancements in perioperative systemic chemotherapy, cytoreductive surgery (CRS), and hyperthermic intraperitoneal chemotherapy (HIPEC) have significantly improved clinical outcomes. PSMs predominantly result from the spread of intra-abdominal neoplasia, which then form secondary peritoneal metastases. Colorectal, ovarian, and gastric cancers are the most common contributors. Despite diverse primary origins, the uniqueness of the peritoneum microenvironment shapes the common features of PSMs. Peritoneal metastization involves complex interactions between tumour cells and the peritoneal microenvironment. Fibroblasts play a crucial role, contributing to tumour development, progression, and therapy resistance. Peritoneal metastasis-associated fibroblasts (MAFs) in PSMs exhibit high heterogeneity. Single-cell RNA sequencing technology has revealed that immune-regulatory cancer-associated fibroblasts (iCAFs) seem to be the most prevalent subtype in PSMs. In addition, other major subtypes as myofibroblastic CAFs (myCAFs) and matrix CAFs (mCAFs) were frequently observed across PSMs studies. Peritoneal MAFs are suggested to originate from mesothelial cells, submesothelial fibroblasts, pericytes, endothelial cells, and omental-resident cells. This plasticity and heterogeneity of CAFs contribute to the complex microenvironment in PSMs, impacting treatment responses. Understanding these interactions is crucial for developing targeted and local therapies to improve PSMs patient outcomes.
Collapse
Affiliation(s)
- Cristiano Ramos
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Vasileios Gerakopoulos
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Rudolf Oehler
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
2
|
Janani M, Poorkhani A, Amiriani T, Donyadideh G, Ahmadi F, Jorjanisorkhankalateh Y, Beheshti-Nia F, Kalaei Z, Roudbaraki M, Soltani M, Khori V, Alizadeh AM. Association of future cancer metastases with fibroblast activation protein-α: a systematic review and meta-analysis. Front Oncol 2024; 14:1339050. [PMID: 38751814 PMCID: PMC11094201 DOI: 10.3389/fonc.2024.1339050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/04/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction Fibroblast activation protein-α (FAP-α) is a vital surface marker of cancer-associated fibroblasts, and its high expression is associated with a higher tumor grade and metastasis. A systematic review and a meta-analysis were performed to associate future metastasis with FAP-α expression in cancer. Methods In our meta-analysis, relevant studies published before 20 February 2024 were systematically searched through online databases that included PubMed, Scopus, and Web of Science. The association between FAP-α expression and metastasis, including distant metastasis, lymph node metastasis, blood vessel invasion, vascular invasion, and neural invasion, was evaluated. A pooled odds ratio (OR) with 95% confidence intervals (CI) was reported as the measure of association. Results A total of 28meta-analysis. The random-effects model for five parameters showed that a high FAP-α expression was associated with blood vessel invasion (OR: 3.04, 95% CI: 1.54-5.99, I 2 = 63%, P = 0.001), lymphovascular invasion (OR: 3.56, 95% CI: 2.14-5.93, I 2 = 0.00%, P < 0.001), lymph node metastasis (OR: 2.73, 95% CI: 1.96-3.81, I 2 = 65%, P < 0.001), and distant metastasis (OR: 2.59; 95% CI: 1.16-5.79, I 2 = 81%, P < 0.001). However, our analysis showed no statistically significant association between high FAP-α expression and neural invasion (OR: 1.57, 95% CI: 0.84-2.93, I 2 = 38%, P = 0.161). Conclusions This meta-analysis indicated that cancer cells with a high FAP-α expression have a higher risk of metastasis than those with a low FAP-α expression. These findings support the potential importance of FAP-α as a biomarker for cancer metastasis prediction.
Collapse
Affiliation(s)
- Majid Janani
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhoushang Poorkhani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Taghi Amiriani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ghazaleh Donyadideh
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farahnazsadat Ahmadi
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Fereshteh Beheshti-Nia
- Department of Epidemiology and Biostatistics, School of Public Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Kalaei
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Morad Roudbaraki
- Laboratory of Cell Physiology, Inserm U1003, University of Lille, Villeneuve d’Ascq, France
| | - Mahsa Soltani
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Mohammad Alizadeh
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Gehris J, Ervin C, Hawkins C, Womack S, Churillo AM, Doyle J, Sinusas AJ, Spinale FG. Fibroblast activation protein: Pivoting cancer/chemotherapeutic insight towards heart failure. Biochem Pharmacol 2024; 219:115914. [PMID: 37956895 PMCID: PMC10824141 DOI: 10.1016/j.bcp.2023.115914] [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: 08/25/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023]
Abstract
An important mechanism for cancer progression is degradation of the extracellular matrix (ECM) which is accompanied by the emergence and proliferation of an activated fibroblast, termed the cancer associated fibroblast (CAF). More specifically, an enzyme pathway identified to be amplified with local cancer progression and proliferation of the CAF, is fibroblast activation protein (FAP). The development and progression of heart failure (HF) irrespective of the etiology is associated with left ventricular (LV) remodeling and changes in ECM structure and function. As with cancer, HF progression is associated with a change in LV myocardial fibroblast growth and function, and expresses a protein signature not dissimilar to the CAF. The overall goal of this review is to put forward the postulate that scientific discoveries regarding FAP in cancer as well as the development of specific chemotherapeutics could be pivoted to target the emergence of FAP in the activated fibroblast subtype and thus hold translationally relevant diagnostic and therapeutic targets in HF.
Collapse
Affiliation(s)
- John Gehris
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Charlie Ervin
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Charlotte Hawkins
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Sydney Womack
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Amelia M Churillo
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Jonathan Doyle
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Albert J Sinusas
- Yale University Cardiovascular Imaging Center, New Haven CT, United States
| | - Francis G Spinale
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States.
| |
Collapse
|
4
|
Pascual-Antón L, Sandoval P, González-Mateo GT, Kopytina V, Tomero-Sanz H, Arriero-País EM, Jiménez-Heffernan JA, Fabre M, Egaña I, Ferrer C, Simón L, González-Cortijo L, Sainz de la Cuesta R, López-Cabrera M. Targeting carcinoma-associated mesothelial cells with antibody-drug conjugates in ovarian carcinomatosis. J Pathol 2023; 261:238-251. [PMID: 37555348 DOI: 10.1002/path.6170] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/14/2023] [Accepted: 06/22/2023] [Indexed: 08/10/2023]
Abstract
Ovarian carcinomatosis is characterized by the accumulation of carcinoma-associated mesothelial cells (CAMs) in the peritoneal stroma and mainly originates through a mesothelial-to-mesenchymal transition (MMT) process. MMT has been proposed as a therapeutic target for peritoneal metastasis. Most ovarian cancer (OC) patients present at diagnosis with peritoneal seeding, which makes tumor progression control difficult by MMT modulation. An alternative approach is to use antibody-drug conjugates (ADCs) targeted directly to attack CAMs. This strategy could represent the cornerstone of precision-based medicine for peritoneal carcinomatosis. Here, we performed complete transcriptome analyses of ascitic fluid-isolated CAMs in advanced OC patients with primary-, high-, and low-grade, serous subtypes and following neoadjuvant chemotherapy. Our findings suggest that both cancer biological aggressiveness and chemotherapy-induced tumor mass reduction reflect the MMT-associated changes that take place in the tumor surrounding microenvironment. Accordingly, MMT-related genes, including fibroblast activation protein (FAP), mannose receptor C type 2 (MRC2), interleukin-11 receptor alpha (IL11RA), myristoylated alanine-rich C-kinase substrate (MARCKS), and sulfatase-1 (SULF1), were identified as specific actionable targets in CAMs of OC patients, which is a crucial step in the de novo design of ADCs. These cell surface target receptors were also validated in peritoneal CAMs of colorectal cancer peritoneal implants, indicating that ADC-based treatment could extend to other abdominal tumors that show peritoneal colonization. As proof of concept, a FAP-targeted ADC reduced tumor growth in an OC xenograft mouse model with peritoneal metastasis-associated fibroblasts. In summary, we propose MMT as a potential source of ADC-based therapeutic targets for peritoneal carcinomatosis. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Lucía Pascual-Antón
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Pilar Sandoval
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Guadalupe T González-Mateo
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Valeria Kopytina
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Henar Tomero-Sanz
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Eva María Arriero-País
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | | | | | | | | | | | | | | | - Manuel López-Cabrera
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| |
Collapse
|
5
|
Ji D, Jia J, Cui X, Li Z, Wu A. FAP promotes metastasis and chemoresistance via regulating YAP1 and macrophages in mucinous colorectal adenocarcinoma. iScience 2023; 26:106600. [PMID: 37213233 PMCID: PMC10196996 DOI: 10.1016/j.isci.2023.106600] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 02/16/2023] [Accepted: 03/31/2023] [Indexed: 05/23/2023] Open
Abstract
Mucinous colorectal adenocarcinoma (MC) is less likely to respond to chemotherapy and is associated with poorer prognosis compared with non-MC (NMC). Fibroblast activation protein (FAP) was found and validated to be upregulated in MC patients and was negatively correlated with prognosis and therapeutic outcomes in colorectal cancer (CRC) patients who were treated with adjuvant chemotherapy. Overexpression of FAP promoted CRC cell growth, invasion and metastasis, and enhanced chemoresistance. Myosin phosphatase Rho-interacting protein (MPRIP) was identified as a direct interacting protein of FAP. FAP may influence the efficiency of chemotherapy and prognosis by promoting the crucial functions of CRC and inducing tumor-associated macrophages (TAMs) recruitment and M2 polarization through regulating theRas Homolog Family Member/Hippo/Yes-associated protein (Rho/Hippo/YAP) signaling pathway. Knockdown of FAP could reverse tumorigenicity and chemoresistance in CRC cells. Thus, FAP may serve as a marker for prognosis and therapeutic outcome, as well as a potential therapeutic target to overcome chemoresistance in MC patients.
Collapse
Affiliation(s)
- Dengbo Ji
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Jinying Jia
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Xinxin Cui
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Zhaowei Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Aiwen Wu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China
- Corresponding author
| |
Collapse
|
6
|
Kalaei Z, Manafi-Farid R, Rashidi B, Kiani FK, Zarei A, Fathi M, Jadidi-Niaragh F. The Prognostic and therapeutic value and clinical implications of fibroblast activation protein-α as a novel biomarker in colorectal cancer. Cell Commun Signal 2023; 21:139. [PMID: 37316886 DOI: 10.1186/s12964-023-01151-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/28/2023] [Indexed: 06/16/2023] Open
Abstract
The identification of contributing factors leading to the development of Colorectal Cancer (CRC), as the third fatal malignancy, is crucial. Today, the tumor microenvironment has been shown to play a key role in CRC progression. Fibroblast-Activation Protein-α (FAP) is a type II transmembrane cell surface proteinase expressed on the surface of cancer-associated fibroblasts in tumor stroma. As an enzyme, FAP has di- and endoprolylpeptidase, endoprotease, and gelatinase/collagenase activities in the Tumor Microenvironment (TME). According to recent reports, FAP overexpression in CRC contributes to adverse clinical outcomes such as increased lymph node metastasis, tumor recurrence, and angiogenesis, as well as decreased overall survival. In this review, studies about the expression level of FAP and its associations with CRC patients' prognosis are reviewed. High expression levels of FAP and its association with clinicopathological factors have made as a potential target. In many studies, FAP has been evaluated as a therapeutic target and diagnostic factor into which the current review tries to provide a comprehensive insight. Video Abstract.
Collapse
Affiliation(s)
- Zahra Kalaei
- Department of Biology, Faculty of Natural Sciences, Tabriz University, Tabriz, Iran
| | - Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bentolhoda Rashidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Karoon Kiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asieh Zarei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehrdad Fathi
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Miao Y, Deng Y, Liu J, Wang J, Hu B, Hao S, Wang H, Zhang Z, Jin Z, Zhang Y, Li C, Zhang P, Wan H, Zhang S, Feng J, Ji N. Anti-cancer effect of targeting fibroblast activation protein alpha in glioblastoma through remodeling macrophage phenotype and suppressing tumor progression. CNS Neurosci Ther 2022; 29:878-892. [PMID: 36382346 PMCID: PMC9928553 DOI: 10.1111/cns.14024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 09/12/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Glioblastoma (GBM) is the most malignant form of glioma and has a poor median survival time. Fibroblast activation protein alpha (FAP) is a dual-specificity serine protease that is strongly associated with the development and progression of human carcinomas. However, relatively little is known about the function of FAP and its potential as a therapeutic target in GBMs. AIMS In this study, we aimed to explore the role of FAP in GBM through a series of experiments and to evaluate the therapeutic effect of PT100, a small molecule inhibitor of FAP, on GBM. RESULTS Increased FAP expression was associated with poor survival in glioma. In vitro, FAP knockdown inhibited the process of EMT and caused a decrease in the number of M2 macrophages. In vivo, PT100 was confirmed to suppress the progression of GBMs significantly. CONCLUSIONS FAP could serve as a biomarker and novel therapeutic target for the treatment of GBM and that PT100 is a promising drug for the treatment of GBM.
Collapse
Affiliation(s)
- Yazhou Miao
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,National Clinical Research Center for Neurological Diseases (China)BeijingChina
| | - Yuxuan Deng
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,National Clinical Research Center for Neurological Diseases (China)BeijingChina
| | - Jinqiu Liu
- Beijing Neurosurgical InstituteCapital Medical UniversityFengtai, BeijingChina
| | - Jing Wang
- Beijing Neurosurgical InstituteCapital Medical UniversityFengtai, BeijingChina
| | - Boyi Hu
- Beijing Neurosurgical InstituteCapital Medical UniversityFengtai, BeijingChina
| | - Shuyu Hao
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,National Clinical Research Center for Neurological Diseases (China)BeijingChina
| | - Herui Wang
- Neuro‐Oncology Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Zhe Zhang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,National Clinical Research Center for Neurological Diseases (China)BeijingChina
| | - Zeping Jin
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,National Clinical Research Center for Neurological Diseases (China)BeijingChina
| | - Yang Zhang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,National Clinical Research Center for Neurological Diseases (China)BeijingChina
| | - Chunzhao Li
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,National Clinical Research Center for Neurological Diseases (China)BeijingChina
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,National Clinical Research Center for Neurological Diseases (China)BeijingChina
| | - Hong Wan
- Beijing Neurosurgical InstituteCapital Medical UniversityFengtai, BeijingChina
| | - Shaodong Zhang
- Beijing Neurosurgical InstituteCapital Medical UniversityFengtai, BeijingChina
| | - Jie Feng
- Beijing Neurosurgical InstituteCapital Medical UniversityFengtai, BeijingChina,Beijing Cancer Institute, Beijing Institute for Brain DisordersCapital Medical UniversityBeijingChina
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,National Clinical Research Center for Neurological Diseases (China)BeijingChina,Beijing Advanced Innovation Center for Big Data‐Based Precision Medicine, School of Engineering MedicineBeihang UniversityBeijingChina
| |
Collapse
|
8
|
Zhang L, Ying W, Sheng Z, Lv L, Gao J, Xue Y, Liu L. Bioluminescence imaging of fibroblast activation protein-alpha in vivo and human plasma with highly sensitive probe. Anal Biochem 2022; 655:114859. [PMID: 35988797 DOI: 10.1016/j.ab.2022.114859] [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: 07/25/2022] [Accepted: 08/09/2022] [Indexed: 11/01/2022]
Abstract
Fibroblast activation protein-alpha (FAPα) has emerged as a biomarker of tumor stromal fibroblasts. FAP was overexpressed in stromal fibroblasts of human malignancies and positively correlated with the depth of tumor invasion, lymphatic metastasis, distant metastases, high TNM stage and poor prognosis. However, the circulating FAP levels in the plasma of gastric cancer patients and the relationship between FAP levels and gastric cancer remain unknown. Moreover, probes with super selectivity, extremely high sensitivity, and excellent performance in quantitative detection are still lacking. Herein, we developed the bioluminescent probe BL-FAP for sensitive detection and imaging of endogenous FAP in gastric cancer cells and tissues and plasma from gastric cancer patients. The probe exhibited the high signal-to-noise ratio (15000∼fold), the excellent selectivity (FAP/DPP IV ratio and FAP/PREP ratio = 50000∼ fold), and the high sensitivity (18.1 pg/mL). BL-FAP facilitates monitoring of endogenous FAP in living cells and nude mice bearing MGC-803-luc tumors. More importantly, this probe was successfully applied to the measurement of FAP activity levels in plasma from gastric cancer patients for the first time. A significant enhancement in FAP levels was observed in patients with gastric cancer, suggesting that the FAP level may be a potential diagnostic parameter for gastric cancer.
Collapse
Affiliation(s)
- Ling Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221002, PR China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, 210023, PR China.
| | - Weiwu Ying
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221002, PR China
| | - Zhijia Sheng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221002, PR China
| | - Li Lv
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221002, PR China
| | - Jian Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221002, PR China
| | - Yunsheng Xue
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221002, PR China
| | - Ling Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221002, PR China
| |
Collapse
|
9
|
Uno K, Iyoshi S, Yoshihara M, Kitami K, Mogi K, Fujimoto H, Sugiyama M, Koya Y, Yamakita Y, Nawa A, Kanayama T, Tomita H, Enomoto A, Kajiyama H. Metastatic Voyage of Ovarian Cancer Cells in Ascites with the Assistance of Various Cellular Components. Int J Mol Sci 2022; 23:4383. [PMID: 35457198 PMCID: PMC9031612 DOI: 10.3390/ijms23084383] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/16/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy and has a unique metastatic route using ascites, known as the transcoelomic root. However, studies on ascites and contained cellular components have not yet been sufficiently clarified. In this review, we focus on the significance of accumulating ascites, contained EOC cells in the form of spheroids, and interaction with non-malignant host cells. To become resistant against anoikis, EOC cells form spheroids in ascites, where epithelial-to-mesenchymal transition stimulated by transforming growth factor-β can be a key pathway. As spheroids form, EOC cells are also gaining the ability to attach and invade the peritoneum to induce intraperitoneal metastasis, as well as resistance to conventional chemotherapy. Recently, accumulating evidence suggests that EOC spheroids in ascites are composed of not only cancer cells, but also non-malignant cells existing with higher abundance than EOC cells in ascites, including macrophages, mesothelial cells, and lymphocytes. Moreover, hetero-cellular spheroids are demonstrated to form more aggregated spheroids and have higher adhesion ability for the mesothelial layer. To improve the poor prognosis, we need to elucidate the mechanisms of spheroid formation and interactions with non-malignant cells in ascites that are a unique tumor microenvironment for EOC.
Collapse
Affiliation(s)
- Kaname Uno
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 223-62 Lund, Sweden
| | - Shohei Iyoshi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| | - Kazuhisa Kitami
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| | - Kazumasa Mogi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| | - Hiroki Fujimoto
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Discipline of Obstetrics and Gynecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia
| | - Mai Sugiyama
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Yoshihiro Koya
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Yoshihiko Yamakita
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Akihiro Nawa
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Tomohiro Kanayama
- Department of Tumor Pathology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (T.K.); (H.T.)
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (T.K.); (H.T.)
| | - Atsushi Enomoto
- Department of Pathology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan;
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| |
Collapse
|
10
|
Demuytere J, Ceelen W, Van Dorpe J, Hoorens A. The role of the peritoneal microenvironment in the pathogenesis of colorectal peritoneal carcinomatosis. Exp Mol Pathol 2020; 115:104442. [PMID: 32305340 DOI: 10.1016/j.yexmp.2020.104442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 01/06/2023]
Abstract
Recent insights have implicated mesothelial-to-mesenchymal transition (MMT) as a mechanism by which mesothelial cells can transdifferentiate into cancer-associated fibroblasts (CAFs) in several cancers metastasizing to the peritoneum. However, this was not evaluated extensively in colorectal cancer. We examined the presumed mesothelial origin of CAFs in three types of colorectal carcinoma: conventional type adenocarcinoma, mucinous carcinoma and signet ring cell carcinoma. We evaluated the expression of mesothelial, mesenchymal, angiogenesis and colorectal cancer-related markers in peritoneal samples of twelve colorectal cancer patients with peritoneal carcinomatosis and four control patients by immunohistochemistry. We observed morphological and immunohistochemical changes in the vicinity of tumor implants in all studied colorectal cancer types. Mesothelial cells acquired a spindle-shaped myofibroblast-like morphology, lost expression of mesothelial markers, and gained expression of mesenchymal markers. Analysis of consecutive tissue sections and double staining for mesothelial and mesenchymal markers revealed overlap in expression of mesothelial and CAF markers. These findings are highly suggestive of a mesothelial origin of CAFs in peritoneal carcinomatosis in colorectal cancer. Interfering with the process of MMT might be a valuable approach in treating and preventing peritoneal carcinomatosis. Differences observed between colorectal cancer types suggest that one single strategy might not be applicable.
Collapse
Affiliation(s)
- Jesse Demuytere
- Laboratory of Experimental Surgery, Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Wim Ceelen
- Laboratory of Experimental Surgery, Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Anne Hoorens
- Department of Pathology, Ghent University Hospital, Ghent, Belgium..
| |
Collapse
|
11
|
Puré E, Blomberg R. Pro-tumorigenic roles of fibroblast activation protein in cancer: back to the basics. Oncogene 2018; 37:4343-4357. [PMID: 29720723 PMCID: PMC6092565 DOI: 10.1038/s41388-018-0275-3] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/23/2018] [Accepted: 01/29/2018] [Indexed: 02/06/2023]
Abstract
Fibroblast activation protein (FAP) is a cell-surface serine protease that acts on various hormones and extracellular matrix components. FAP is highly upregulated in a wide variety of cancers, and is often used as a marker for pro-tumorigenic stroma. It has also been proposed as a molecular target of cancer therapies, and, especially in recent years, a great deal of research has gone into design and testing of diverse FAP-targeted treatments. Yet despite this growing field of research, our knowledge of FAP's basic biology and functional roles in various cancers has lagged behind its use as a tumor-stromal marker. In this review, we summarize and analyze recent advances in understanding the functions of FAP in cancer, most notably its prognostic value in various tumor types, cellular effects on various cell types, and potential as a therapeutic target. We highlight outstanding questions in the field, the answers to which could shape preclinical and clinical studies of FAP.
Collapse
Affiliation(s)
- Ellen Puré
- University of Pennsylvania, Philadelphia, PA, USA.
| | | |
Collapse
|
12
|
Mikuła-Pietrasik J, Uruski P, Tykarski A, Książek K. The peritoneal "soil" for a cancerous "seed": a comprehensive review of the pathogenesis of intraperitoneal cancer metastases. Cell Mol Life Sci 2018; 75:509-525. [PMID: 28956065 PMCID: PMC5765197 DOI: 10.1007/s00018-017-2663-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/29/2017] [Accepted: 09/20/2017] [Indexed: 01/02/2023]
Abstract
Various types of tumors, particularly those originating from the ovary and gastrointestinal tract, display a strong predilection for the peritoneal cavity as the site of metastasis. The intraperitoneal spread of a malignancy is orchestrated by a reciprocal interplay between invading cancer cells and resident normal peritoneal cells. In this review, we address the current state-of-art regarding colonization of the peritoneal cavity by ovarian, colorectal, pancreatic, and gastric tumors. Particular attention is paid to the pro-tumoral role of various kinds of peritoneal cells, including mesothelial cells, fibroblasts, adipocytes, macrophages, the vascular endothelium, and hospicells. Anatomo-histological considerations on the pro-metastatic environment of the peritoneal cavity are presented in the broader context of organ-specific development of distal metastases in accordance with Paget's "seed and soil" theory of tumorigenesis. The activity of normal peritoneal cells during pivotal elements of cancer progression, i.e., adhesion, migration, invasion, proliferation, EMT, and angiogenesis, is discussed from the perspective of well-defined general knowledge on a hospitable tumor microenvironment created by the cellular elements of reactive stroma, such as cancer-associated fibroblasts and macrophages. Finally, the paper addresses the unique features of the peritoneal cavity that predispose this body compartment to be a niche for cancer metastases, presents issues that are topics of an ongoing debate, and points to areas that still require further in-depth investigations.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland
| | - Paweł Uruski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland
| | - Krzysztof Książek
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland.
| |
Collapse
|
13
|
Lv ZD, Zhao WJ, Jin LY, Wang WJ, Dong Q, Li N, Xu HM, Wang HB. Blocking TGF-β1 by P17 peptides attenuates gastric cancer cell induced peritoneal fibrosis and prevents peritoneal dissemination in vitro and in vivo. Biomed Pharmacother 2017; 88:27-33. [PMID: 28092842 DOI: 10.1016/j.biopha.2017.01.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 12/25/2016] [Accepted: 01/05/2017] [Indexed: 12/20/2022] Open
Abstract
Our previous study demonstrated that the peritoneal stroma environment favors proliferation of tumor cells by serving as a rich source of growth factors and chemokines known to be involved in tumor metastasis. In this study, we investigated the interaction between gastric cancer cells and peritoneal mesothelial cells, and determined the effects of TGF-β1 in this processing. Human peritoneal tissues and peritoneal wash fluid were obtained, which examined by hematoxylin and eosin staining or ELISA for measurements of TGF-β1 levels. The peritoneal mesothelial cells were co-incubated with the supernatants of gastric cancer, the expression of TGF-β1, collagen and fibronectin was observed by ELISA and western blot. We then investigated the effects of serum-free conditioned media from HSC-39 gastric cancer cells on the peritoneum of nude mice, and the effects of peritoneal fibrosis on the development of peritoneal metastasis in vivo. The peritoneum from gastric patients were thickened and contained extensive fibrosis. After co-culture both gastric tumor cells and mesothelial cells, we found that TGF-β1 expression was greatly increased in the co-culture system compared to individual culture condition. Serum-free Conditioned Media from HSC-39 was able to induce extracellular matrix expression in vitro and in vivo, and tumorigenicity in mice with peritoneal fibrosis was greater than in mice with normal peritoneum, while blocking TGF-β1 by peptide P17 can partially inhibit these effects. In conclusion, these results indicated that the interaction of gastric cancer with peritoneal fibrosis and determined that TGF-β1 plays a key role in induction of peritoneal fibrosis, which in turn affected dissemination of gastric cancer.
Collapse
Affiliation(s)
- Zhi-Dong Lv
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China; Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang 110001, PR China
| | - Wei-Jun Zhao
- Department of General Surgery, The Affiliated Hospital of Chifeng University, Chifeng 024000, PR China
| | - Li-Ying Jin
- Cerebrovascular Disease Research Institute and (e)Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Wen-Juan Wang
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Qian Dong
- Departments of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Na Li
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Hui-Mian Xu
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang 110001, PR China.
| | - Hai-Bo Wang
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China.
| |
Collapse
|
14
|
Endoglin overexpression mediates gastric cancer peritoneal dissemination by inducing mesothelial cell senescence. Hum Pathol 2016; 51:114-23. [PMID: 27067789 DOI: 10.1016/j.humpath.2015.12.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 12/22/2015] [Accepted: 12/23/2015] [Indexed: 01/08/2023]
Abstract
Peritoneal dissemination (PD), which is highly frequent in gastric cancer (GC) patients, is the main cause of death in advanced GC. Senescence of human peritoneal mesothelial cells (HPMC) may contribute to GC peritoneal dissemination (GCPD). In this study of 126 patients, we investigated the association between Endoglin expression in GC peritoneum and the clinicopathological features. The prognosis of patients was evaluated according to Endoglin and ID1 expression. In vitro, GC cell (GCC)-HPMC coculture was established. Endoglin and ID1 expression was evaluated by Western blot. Cell cycle and HPMC senescence were analyzed after harvesting HPMC from the coculture. GCC adhesion and invasion to HPMC were also assayed. Our results showed that positive staining of Endoglin (38%) was associated with a higher TNM stage and higher incidence of GCPD (both P < .05). Kaplan-Meier analysis showed that the patients who were Endoglin positive had a shorter survival time compared with Endoglin-negative patients (P = .02). Using the HPMC and GCC adherence and invasion assay, we demonstrated that transforming growth factor beta 1 (TGF-β)1-induced HPMC senescence was attenuated by silencing the Endoglin expression, which also prevented GCC attachment and invasion. Our study indicated a positive correlation between Endoglin overexpression and GCPD. Up-regulated Endoglin expression induced HPMC senescence via TGF-β1 pathway. The findings suggest that Endoglin-induced HPMC senescence may contribute to peritoneal dissemination of GCCs.
Collapse
|
15
|
The Mesothelial Origin of Carcinoma Associated-Fibroblasts in Peritoneal Metastasis. Cancers (Basel) 2015; 7:1994-2011. [PMID: 26426054 PMCID: PMC4695872 DOI: 10.3390/cancers7040872] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/14/2015] [Accepted: 09/23/2015] [Indexed: 01/15/2023] Open
Abstract
Solid tumors are complex and unstructured organs that, in addition to cancer cells, also contain other cell types. Carcinoma-associated fibroblasts (CAFs) represent an important population in the tumor microenviroment and participate in several stages of tumor progression, including cancer cell migration/invasion and metastasis. During peritoneal metastasis, cancer cells detach from the primary tumor, such as ovarian or gastrointestinal, disseminate through the peritoneal fluid and colonize the peritoneum. Tumor cells metastasize by attaching to and invading through the mesothelial cell (MC) monolayer that lines the peritoneal cavity, then colonizing the submesothelial compact zone where CAFs accumulate. CAFs may derive from different sources depending on the surrounding metastatic niche. In peritoneal metastasis, a sizeable subpopulation of CAFs originates from MCs through a mesothelial-to-mesenchymal transition (MMT), which promotes adhesion, invasion, vascularization and subsequent tumor growth. The bidirectional communication between cancer cells and MC-derived CAFs via secretion of a wide range of cytokines, growth factors and extracellular matrix components seems to be crucial for the establishment and progression of the metastasis in the peritoneum. This manuscript provides a comprehensive review of novel advances in understanding how peritoneal CAFs provide cancer cells with a supportive microenvironment, as well as the development of future therapeutic approaches by interfering with the MMT in the peritoneum.
Collapse
|