1
|
van der Heide CD, Campeiro JD, Ruigrok EAM, van den Brink L, Ponnala S, Hillier SM, Dalm SU. In vitro and ex vivo evaluation of preclinical models for FAP-targeted theranostics: differences and relevance for radiotracer evaluation. EJNMMI Res 2024; 14:125. [PMID: 39718718 DOI: 10.1186/s13550-024-01191-6] [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: 10/09/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Fibroblast activation protein (FAP) is an attractive target for cancer theranostics. Although FAP-targeted nuclear imaging demonstrated promising clinical results, only sub-optimal results are reported for targeted radionuclide therapy (TRT). Preclinical research is crucial in selecting promising FAP-targeted radiopharmaceuticals and for obtaining an increased understanding of factors essential for FAP-TRT improvement. FAP is mainly expressed by cancer-associated fibroblasts in the tumor stroma and less on cancer cells themselves. Therefore, other (complex) factors impact FAP-TRT efficacy compared to currently clinically applied TRT strategies. For accurate evaluation of these aspects, selection of a representative preclinical model is important. Currently mainly human cancer cell lines transduced to (over)express FAP are applied, lacking clinical representation. It is unclear how these and more physiological FAP-expressing models compare to each other, and whether/how the model influences the study outcome. We aimed to address this by comparing FAP tracer behavior in FAP-transduced HT1080-huFAP and HEK293-huFAP cells, and endogenous FAP-expressing U-87 MG cancer cells and PS-1 pancreatic stellate cells. [111In]In-FAPI-46 and a fluorescent FAP-targeted tracer (RTX-1370S) were used to compare tracer binding/uptake and localization in vitro and ex vivo. Additionally, FAP expression was determined with RT-qPCR and anti-FAP IHC. RESULTS Although FAP expression was highest in HEK293-huFAP cells and cell line derived xenografts, this did not result in the highest tracer uptake. [111In]In-FAPI-46 uptake was highest in HT1080-huFAP, closely followed by HEK293-huFAP, and a 6-10-fold lower uptake for U-87 MG and PS-1 cells. However, ex vivo U-87 MG xenografts only showed a 2-fold lower binding compared to HT1080-huFAP and HEK293-huFAP xenografts, mainly because the cell line attracts murine fibroblasts as demonstrated in our RT-qPCR and IHC studies. CONCLUSIONS The interaction between FAP and FAP-targeted tracers differs between models, indicating the need for appropriate model selection and that comparing results across studies using different models is difficult.
Collapse
Affiliation(s)
- Circe D van der Heide
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, GD, 3015, The Netherlands
| | - Joana D Campeiro
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, GD, 3015, The Netherlands
| | - Eline A M Ruigrok
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, GD, 3015, The Netherlands
| | - Lilian van den Brink
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, GD, 3015, The Netherlands
| | | | | | - Simone U Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Centre Rotterdam, Rotterdam, GD, 3015, The Netherlands.
| |
Collapse
|
2
|
Zhao Y, Li Y, Zou J, Guo T, Zhong Z, Li Y, Chen S, Li J, Huang K, Lian G, Huang Y. Low-dose arsenic trioxide inhibits pancreatic stellate cell activation via LOXL3 expression to enhance immunotherapy in pancreatic cancer. Br J Cancer 2024; 131:1928-1941. [PMID: 39501090 PMCID: PMC11628614 DOI: 10.1038/s41416-024-02880-8] [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: 01/31/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is characterized by abnormally fibrotic mesenchyme, which notably influences on the effectiveness of immunotherapy. Low-dose arsenic trioxide (ATO, 1.0 μM) can inhibit the activation of pancreatic stellate cells (PSCs) and affect fibrosis, which is a potential strategy for enhancing the sensitivity to immunotherapy. METHODS Extracellular matrix (ECM) models were employed to assess the regulatory effects of ATO on ECM and peripheral blood mononuclear cells. Orthotopic C57BL/6J models were utilized to evaluate the influence of ATO on CD8+T cell infiltration and immunotherapy in PC. Additionally, nanomaterials loaded with ATO designed to specifically target PSCs (scAbFAP-α-HMSNs-PAA-ATO) were produced to enhance targeting effects of ATO. RESULTS Low-dose ATO (1.0 μM) suppressed PSCs activation, exhibiting potential for synergistic immunotherapy. Under low-dose ATO intervention, ECM underwent remodeling, leading to increases in CD8+T cell infiltration, thereby enhancing anti-PD-L1 therapy effect. We further demonstrated that low-dose ATO remodeled ECM by regulating the expression of LOXL3 in PSCs. scAbFAP-α-HMSNs-PAA-ATO exhibited improved targeting capabilities, and enhanced capacity to inhibit fibrosis and sensitize immunotherapy. CONCLUSIONS Our research reveals that low-dose ATO, by regulating LOXL3, remodels the ECM and enhances CD8+T cell infiltration, thus sensitizing the efficacy of immunotherapy, which provides a novel strategy for comprehensive treatment to PC.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yunlong Li
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jinmao Zou
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tairan Guo
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ziyi Zhong
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yaqing Li
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shaojie Chen
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiajia Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Kaihong Huang
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Guoda Lian
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Yuzhou Huang
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
3
|
Beer P, Pauli C, Haberecker M, Grest P, Beebe E, Fuchs D, Markkanen E, Krudewig C, Nolff MC. Cross-species evaluation of fibroblast activation protein alpha as potential imaging target for soft tissue sarcoma: a comparative immunohistochemical study in humans, dogs, and cats. Front Oncol 2023; 13:1210004. [PMID: 37727209 PMCID: PMC10505752 DOI: 10.3389/fonc.2023.1210004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/09/2023] [Indexed: 09/21/2023] Open
Abstract
Introduction Complete surgical tumor resection is paramount in the management of soft tissue sarcoma (STS) in humans, dogs, and cats alike. Near-infrared targeted tracers for fluorescence-guided surgery (FGS) could facilitate intraoperative visualization of the tumor and improve resection accuracy. Target identification is complicated in STS due to the rarity and heterogeneity of the disease. This study aims to validate the expression of fibroblast activation protein alpha (FAP) in selected human, canine, and feline STS subtypes to assess the value of FAP as a target for FGS and to validate companion animals as a translational model. Methods Formalin-fixed and paraffin-embedded tissue samples from 53 canine STSs (perivascular wall tumor (PWT), canine fibrosarcoma (cFS), and STS not further specified (NOS)), 24 feline fibrosarcomas, and 39 human STSs (myxofibrosarcoma, undifferentiated pleomorphic sarcoma, dermatofibrosarcoma protuberans, and malignant peripheral nerve sheath tumor) as well as six canine and seven feline healthy controls and 10 inflamed tissue samples were immunohistochemically stained for their FAP expression. FAP labeling in tumor, peritumoral, healthy skin, and inflamed tissue samples was quantified using a visually assessed semiquantitative expression score and digital image analysis. Target selection criteria (TASC) scoring was subsequently performed as previously described. Results Eighty-five percent (85%) of human (33/39), 76% of canine (40/53), and 92% of feline (22/24) STSs showed FAP positivity in over 10% of the tumor cells. A high expression was determined in 53% canine (28/53), 67% feline (16/24), and 44% human STSs (17/39). The average FAP-labeled area of canine, feline, and human STSs was 31%, 33%, and 42%, respectively (p > 0.8990). The FAP-positive tumor area was larger in STS compared to healthy and peritumoral tissue samples (p < 0.0001). TASC scores were above 18 for all feline and human STS subtypes and canine PWTs but not for canine STS NOS and cFS. Conclusion This study represents the first cross-species target evaluation of FAP for STS. Our results demonstrate that FAP expression is increased in various STS subtypes compared to non-cancerous tissues across species, thereby validating dogs and cats as suitable animal models. Based on a TASC score, FAP could be considered a target for FGS.
Collapse
Affiliation(s)
- Patricia Beer
- Clinic for Small Animal Surgery, Department for Small Animals, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Chantal Pauli
- Department of Pathology and Molecular Pathology, University of Zurich, Zurich, Switzerland
- Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Martina Haberecker
- Department of Pathology and Molecular Pathology, University of Zurich, Zurich, Switzerland
| | - Paula Grest
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Erin Beebe
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Daniel Fuchs
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Christiane Krudewig
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Mirja Christine Nolff
- Clinic for Small Animal Surgery, Department for Small Animals, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
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
|
5
|
Olajubutu O, Ogundipe OD, Adebayo A, Adesina SK. Drug Delivery Strategies for the Treatment of Pancreatic Cancer. Pharmaceutics 2023; 15:pharmaceutics15051318. [PMID: 37242560 DOI: 10.3390/pharmaceutics15051318] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Pancreatic cancer is fast becoming a global menace and it is projected to be the second leading cause of cancer-related death by 2030. Pancreatic adenocarcinomas, which develop in the pancreas' exocrine region, are the predominant type of pancreatic cancer, representing about 95% of total pancreatic tumors. The malignancy progresses asymptomatically, making early diagnosis difficult. It is characterized by excessive production of fibrotic stroma known as desmoplasia, which aids tumor growth and metastatic spread by remodeling the extracellular matrix and releasing tumor growth factors. For decades, immense efforts have been harnessed toward developing more effective drug delivery systems for pancreatic cancer treatment leveraging nanotechnology, immunotherapy, drug conjugates, and combinations of these approaches. However, despite the reported preclinical success of these approaches, no substantial progress has been made clinically and the prognosis for pancreatic cancer is worsening. This review provides insights into challenges associated with the delivery of therapeutics for pancreatic cancer treatment and discusses drug delivery strategies to minimize adverse effects associated with current chemotherapy options and to improve the efficiency of drug treatment.
Collapse
Affiliation(s)
| | - Omotola D Ogundipe
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| | - Amusa Adebayo
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| | - Simeon K Adesina
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| |
Collapse
|
6
|
Cheng CS, Yang PW, Sun Y, Song SL, Chen Z. Fibroblast activation protein-based theranostics in pancreatic cancer. Front Oncol 2022; 12:969731. [PMID: 36263225 PMCID: PMC9574192 DOI: 10.3389/fonc.2022.969731] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Fibroblast activation protein-α (FAP) is a type II transmembrane serine protease that has specific endopeptidase activity. Given its well-established selective expression in the activated stromal fibroblasts of epithelial cancers, although not in quiescent fibroblasts, FAP has received substantial research attention as a diagnostic marker and therapeutic target. Pancreatic cancer is characterized by an abundant fibrotic or desmoplastic stroma, leading to rapid progression, therapeutic resistance, and poor clinical outcomes. Numerous studies have revealed that the abundant expression of FAP in cancer cells, circulating tumor cells, stromal cells, and cancer-associated fibroblasts (CAFs) of pancreatic adenocarcinoma is implicated in diverse cancer-related signaling pathways, contributing to cancer progression, invasion, migration, metastasis, immunosuppression, and resistance to treatment. In this article, we aim to systematically review the recent advances in research on FAP in pancreatic adenocarcinoma, including its utility as a diagnostic marker, therapeutic potential, and correlation with prognosis. We also describe the functional role of FAP-overexpressing stromal cells, particulary CAFs, in tumor immuno- and metabolic microenvironments, and summarize the mechanisms underlying the contribution of FAP-overexpressing CAFs in pancreatic cancer progression and treatment resistance. Furthermore, we discuss whether targeting FAP-overexpressing CAFs could represent a potential therapeutic strategy and describe the development of FAP-targeted probes for diagnostic imaging. Finally, we assess the emerging basic and clinical studies regarding the bench-to-bedside translation of FAP in pancreatic cancer.
Collapse
Affiliation(s)
- Chien-shan Cheng
- Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, China
| | - Pei-wen Yang
- Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun Sun
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
| | - Shao-li Song
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Nuclear Medicine Department, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Zhen Chen
- Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Zhen Chen,
| |
Collapse
|
7
|
Meng Y, Yu J, Zhu M, Zhou J, Li N, Liu F, Zhang H, Fang X, Li J, Feng X, Wang L, Jiang H, Lu J, Shao C, Bian Y. CT radiomics signature: a potential biomarker for fibroblast activation protein expression in patients with pancreatic ductal adenocarcinoma. Abdom Radiol (NY) 2022; 47:2822-2834. [PMID: 35451626 DOI: 10.1007/s00261-022-03512-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE To develop and validate a radiomics model to predict fibroblast activation protein (FAP) expression in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS This retrospective study included consecutive 152 patients with PDAC who underwent MDCT scan and surgical resection from January 2017 to December 2017 (training set) and from January 2018 to April 2018 (validation set). In the training set, 1409 portal radiomic features were extracted from each patient's preoperative imaging. Optimal features were selected using the least absolute shrinkage and selection operator (LASSO) logistic regression algorithm, whereupon the extreme gradient boosting (XGBoost) was developed using the radiomics features. The performance of the XGBoost classifier performance was assessed by its calibration, discrimination, and clinical usefulness. RESULTS The patients were divided into FAP-low (n = 91; 59.87%) and FAP-high (n = 61; 40.13%) groups according to the optimal FAP cutoff (45.71%). Patients in the FAP-low group showed longer survival. The XGBoost classifier comprised 13 selected radiomics features and showed good discrimination in the training set [area under the curve (AUC), 0.97] and the validation set (AUC, 0.75). It also performed well in the calibration test and decision-curve analysis, demonstrating its potential clinical value. CONCLUSIONS The XGBoost classifier based on CT radiomics in the portal venous phase can non-invasively predict FAP expression and may help to improve clinical decision-making in patients with PDAC.
Collapse
Affiliation(s)
- Yinghao Meng
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Department of Radiology, No. 971 Hospital of Navy, Qingdao, Shandong, China
| | - Jieyu Yu
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Mengmeng Zhu
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jian Zhou
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Na Li
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Fang Liu
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hao Zhang
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xu Fang
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jing Li
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaocheng Feng
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Li Wang
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianping Lu
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China.
- Department of Radiology, Changhai Hospital, 168 Changhai Road, Shanghai, 200433, China.
| | - Yun Bian
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China.
- Department of Radiology, Changhai Hospital, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
8
|
van der Heide CD, Dalm SU. Radionuclide imaging and therapy directed towards the tumor microenvironment: a multi-cancer approach for personalized medicine. Eur J Nucl Med Mol Imaging 2022; 49:4616-4641. [PMID: 35788730 PMCID: PMC9606105 DOI: 10.1007/s00259-022-05870-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022]
Abstract
Targeted radionuclide theranostics is becoming more and more prominent in clinical oncology. Currently, most nuclear medicine compounds researched for cancer theranostics are directed towards targets expressed in only a small subset of cancer types, limiting clinical applicability. The identification of cancer-specific targets that are (more) universally expressed will allow more cancer patients to benefit from these personalized nuclear medicine–based interventions. A tumor is not merely a collection of cancer cells, it also comprises supporting stromal cells embedded in an altered extracellular matrix (ECM), together forming the tumor microenvironment (TME). Since the TME is less genetically unstable than cancer cells, and TME phenotypes can be shared between cancer types, it offers targets that are more universally expressed. The TME is characterized by the presence of altered processes such as hypoxia, acidity, and increased metabolism. Next to the ECM, the TME consists of cancer-associated fibroblasts (CAFs), macrophages, endothelial cells forming the neo-vasculature, immune cells, and cancer-associated adipocytes (CAAs). Radioligands directed at the altered processes, the ECM, and the cellular components of the TME have been developed and evaluated in preclinical and clinical studies for targeted radionuclide imaging and/or therapy. In this review, we provide an overview of the TME targets and their corresponding radioligands. In addition, we discuss what developments are needed to further explore the TME as a target for radionuclide theranostics, with the hopes of stimulating the development of novel TME radioligands with multi-cancer, or in some cases even pan-cancer, application.
Collapse
Affiliation(s)
| | - Simone U Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
9
|
Liu SY, Wang H, Nie G. Ultrasensitive Fibroblast Activation Protein-α-Activated Fluorogenic Probe Enables Selective Imaging and Killing of Melanoma In Vivo. ACS Sens 2022; 7:1837-1846. [PMID: 35713201 DOI: 10.1021/acssensors.2c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Melanoma is a malignant cancer with a high risk of metastasis and continued increase in death rates over the past decades, and its prognosis is highly related to the disease's stage, while early detection and treatment of melanoma are significant to the improvement of its therapy outcome. Different from the traditional methods for disease diagnosis, enzyme-activated fluorescent probes were developed rapidly due to their high sensitivity and temporal-spatial ratio and have been widely applied in tumor detection, surgical navigation, and cancer-related research. Fibroblast activation protein-α (FAPα), a serine-type cell surface protease that plays important roles in cell invasion and extracellular matrix degradation, is widely involved in tumor progression such as malignant melanoma, so developing a FAPα activity-based molecular tool would be of great potential for the early diagnosis and therapy of melanoma. However, few fluorescent probes targeting FAPα have been applied in melanoma-related studies, and thus, the construction of FAPα activity-based fluorescent probes for melanoma detection is in urgent need. By incorporating the selective recognition unit with a red-emission fluorophore, cresyl violet, we herein report an ultrasensitive (limit of detection = 5.3 ng/mL) fluorogenic probe for FAPα activity sensing, named CV-FAP; the acquired probe showed a significantly higher binding affinity (15.7-fold) and overall catalytic efficiency (2.6-fold) when compared with those of the best reported FAPα probes. The good performance of CV-FAP made it possible to discriminate malignant melanoma cells and tumor-bearing mice from normal cells and mice with high contrast. More importantly, CV-FAP showed significant antitumor activity toward melanoma in cultured cells and tumor-bearing nude mice (over 95% inhibited tumor growth) with good safety, which made it an ideal theranostic agent for melanoma.
Collapse
Affiliation(s)
- Shi-Yu Liu
- Department of Laboratory Medicine, School of Medicine, Yangtze University, Jingzhou 434023, P. R. China
| | - Huiling Wang
- College of Chemistry, Central China Normal University, 152 Luoyu Road, Wuhan 430079, P. R. China
| | - Gang Nie
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, P. R. China
| |
Collapse
|
10
|
Peng D, He J, Liu H, Cao J, Wang Y, Chen Y. FAPI PET/CT research progress in digestive system tumours. Dig Liver Dis 2022; 54:164-169. [PMID: 34364808 DOI: 10.1016/j.dld.2021.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/24/2022]
Abstract
18F-fluorodeoxyglucose positron emission tomography/computed tomography has been used in clinical practice for many years. This modality is of great value for tumour diagnosis, staging, and efficacy evaluations, but it has many limitations in the diagnosis and treatment of digestive system tumours. Fibroblast activation protein is highly expressed in gastrointestinal tumours. Various isotope-labelled fibroblast activation protein inhibitors are widely used in clinical research. These inhibitors have low background uptake in the brain, liver and oral/pharyngeal mucosa and show good contrast between the tumour and background, which makes up for the lack of fluorodeoxyglucose in the diagnosis of digestive system tumours. It better displays the primary tumours, metastases and regional lymph nodes of digestive system tumours, such as oesophageal cancer, gastric cancer and liver cancer, and also provides a new method for treating these tumours. Based on this background, this article introduces the current research status of fibroblast activation protein inhibitor positron emission tomography/computed tomography in various types of digestive system malignant tumours to provide more valuable information for diagnosing and treating digestive system tumours.
Collapse
Affiliation(s)
- Dengsai Peng
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No 25 TaiPing St, Jiangyang District, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China; Academician (Expert) Workstation of Sichuan Province, 646000, PR China
| | - Jing He
- Department of Ultrasonography, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Hanxiang Liu
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No 25 TaiPing St, Jiangyang District, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China; Academician (Expert) Workstation of Sichuan Province, 646000, PR China
| | - Jianpeng Cao
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No 25 TaiPing St, Jiangyang District, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China; Academician (Expert) Workstation of Sichuan Province, 646000, PR China
| | - Yingwei Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No 25 TaiPing St, Jiangyang District, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China; Academician (Expert) Workstation of Sichuan Province, 646000, PR China
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No 25 TaiPing St, Jiangyang District, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China; Academician (Expert) Workstation of Sichuan Province, 646000, PR China.
| |
Collapse
|
11
|
Zhao Y, Yao H, Yang K, Han S, Chen S, Li Y, Chen S, Huang K, Lian G, Li J. Arsenic Trioxide-loaded nanoparticles Enhance the Chemosensitivity of Gemcitabine in Pancreatic Cancer via Reversal of Pancreatic Stellate Cells Desmoplasia through Targeting AP4/Galectin-1 Pathway. Biomater Sci 2022; 10:5989-6002. [DOI: 10.1039/d2bm01039a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pancreatic stellate cell (PSCs) constitutes the fibrotic tumor microenvironment composed of the stroma matrix, which blocks the penetration of Gemcitabine (GEM) in pancreatic adenocarcinoma (PDAC) and results in chemoresistance. We...
Collapse
|
12
|
Juillerat-Jeanneret L, Tafelmeyer P, Golshayan D. Regulation of Fibroblast Activation Protein-α Expression: Focus on Intracellular Protein Interactions. J Med Chem 2021; 64:14028-14045. [PMID: 34523930 DOI: 10.1021/acs.jmedchem.1c01010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prolyl-specific peptidase fibroblast activation protein-α (FAP-α) is expressed at very low or undetectable levels in nondiseased human tissues but is selectively induced in activated (myo)fibroblasts at sites of tissue remodeling in fibrogenic processes. In normal regenerative processes involving transient fibrosis FAP-α+(myo)fibroblasts disappear from injured tissues, replaced by cells with a normal FAP-α- phenotype. In chronic uncontrolled pathological fibrosis FAP-α+(myo)fibroblasts permanently replace normal tissues. The mechanisms of regulation and elimination of FAP-α expression in(myo)fibroblasts are unknown. According to a yeast two-hybrid screen and protein databanks search, we propose that the intracellular (co)-chaperone BAG6/BAT3 can interact with FAP-α, mediated by the BAG6/BAT3 Pro-rich domain, inducing proteosomal degradation of FAP-α protein under tissue homeostasis. In this Perspective, we discuss our findings in the context of current knowledge on the regulation of FAP-α expression and comment potential therapeutic strategies for uncontrolled fibrosis, including small molecule degraders (PROTACs)-modified FAP-α targeted inhibitors.
Collapse
Affiliation(s)
- Lucienne Juillerat-Jeanneret
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH1011 Lausanne, Switzerland.,University Institute of Pathology, CHUV and UNIL, CH1011 Lausanne, Switzerland
| | - Petra Tafelmeyer
- Hybrigenics Services, Laboratories and Headquarters-Paris, 1 rue Pierre Fontaine, 91000 Evry, France.,Hybrigenics Corporation, Cambridge Innovation Center, 50 Milk Street, Cambridge, Massachusetts 02142, United States
| | - Dela Golshayan
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH1011 Lausanne, Switzerland
| |
Collapse
|
13
|
Szalontai K, Gémes N, Furák J, Varga T, Neuperger P, Balog JÁ, Puskás LG, Szebeni GJ. Chronic Obstructive Pulmonary Disease: Epidemiology, Biomarkers, and Paving the Way to Lung Cancer. J Clin Med 2021; 10:jcm10132889. [PMID: 34209651 PMCID: PMC8268950 DOI: 10.3390/jcm10132889] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 12/16/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD), the frequently fatal pathology of the respiratory tract, accounts for half a billion cases globally. COPD manifests via chronic inflammatory response to irritants, frequently to tobacco smoke. The progression of COPD from early onset to advanced disease leads to the loss of the alveolar wall, pulmonary hypertension, and fibrosis of the respiratory epithelium. Here, we focus on the epidemiology, progression, and biomarkers of COPD with a particular connection to lung cancer. Dissecting the cellular and molecular players in the progression of the disease, we aim to shed light on the role of smoking, which is responsible for the disease, or at least for the more severe symptoms and worse patient outcomes. We summarize the inflammatory conditions, as well as the role of EMT and fibroblasts in establishing a cancer-prone microenvironment, i.e., the soil for ‘COPD-derived’ lung cancer. We highlight that the major health problem of COPD can be alleviated via smoking cessation, early diagnosis, and abandonment of the usage of biomass fuels on a global basis.
Collapse
Affiliation(s)
- Klára Szalontai
- Csongrád County Hospital of Chest Diseases, Alkotmány u. 36., H6772 Deszk, Hungary;
| | - Nikolett Gémes
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
- PhD School in Biology, University of Szeged, H6726 Szeged, Hungary
| | - József Furák
- Department of Surgery, University of Szeged, Semmelweis u. 8., H6725 Szeged, Hungary;
| | - Tünde Varga
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
| | - Patrícia Neuperger
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
- PhD School in Biology, University of Szeged, H6726 Szeged, Hungary
| | - József Á. Balog
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
- PhD School in Biology, University of Szeged, H6726 Szeged, Hungary
| | - László G. Puskás
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
- Avicor Ltd. Alsó Kikötő sor 11/D, H6726 Szeged, Hungary
| | - Gábor J. Szebeni
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, H6726 Szeged, Hungary
- CS-Smartlab Devices Ltd., Ady E. u. 14., H7761 Kozármisleny, Hungary
- Correspondence:
| |
Collapse
|
14
|
Byrling J, Sasor A, Nilsson J, Said Hilmersson K, Andersson R, Andersson B. Expression of fibroblast activation protein and the clinicopathological relevance in distal cholangiocarcinoma. Scand J Gastroenterol 2020; 55:82-89. [PMID: 31917931 DOI: 10.1080/00365521.2019.1708449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Objectives: The current survival of patients with distal cholangiocarcinoma (dCCA) is poor. There is a need to develop new prognostic and predictive biomarkers to improve the survival of patients. Fibroblast activation protein (FAP) expression has been associated with survival in several solid malignancies. The goal of this study was to evaluate the expression pattern and prognostic significance of FAP in dCCA.Materials and methods: FAP expression was examined in 57 resected dCCA specimens and 28 paired lymph node metastasis specimens, as well as 10 benign bile ducts using immunohistochemistry. FAP expression was scored in the epithelial and stromal component of the dCCA specimens. The association between FAP expression and prognosis was evaluated using univariable and multivariable statistical modeling.Results: FAP expression was absent in the benign controls. FAP expression was evident in the epithelial 43 (75%) and stromal compartment 34 (60%) of dCCA. There was no association between epithelial or stromal FAP expression and clinicopathological factors. Epithelial FAP expression (HR 0.4 95% CI 0.20-0.78; p=.007) but not stromal FAP expression was significantly associated with better survival in univariable and multivariable analysis.Conclusions: FAP overexpression is evident in dCCA. There was a positive association between epithelial FAP expression and better survival which merits further evaluation.
Collapse
Affiliation(s)
- Johannes Byrling
- Department of Surgery, Lund University and Skane University Hospital, Lund, Sweden
| | - Agata Sasor
- Department of Pathology, Lund University and Skane University Hospital, Lund, Sweden
| | - Johan Nilsson
- Department of Cardiothoracic Surgery, Lund University and Skane University Hospital, Lund, Sweden
| | | | - Roland Andersson
- Department of Surgery, Lund University and Skane University Hospital, Lund, Sweden
| | - Bodil Andersson
- Department of Surgery, Lund University and Skane University Hospital, Lund, Sweden
| |
Collapse
|
15
|
Wang FT, Sun W, Zhang JT, Fan YZ. Cancer-associated fibroblast regulation of tumor neo-angiogenesis as a therapeutic target in cancer. Oncol Lett 2019; 17:3055-3065. [PMID: 30867734 PMCID: PMC6396119 DOI: 10.3892/ol.2019.9973] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/21/2018] [Indexed: 12/20/2022] Open
Abstract
Adequate blood supply is essential for tumor survival, growth and metastasis. The tumor microenvironment (TME) is dynamic and complex, comprising cancer cells, cancer-associated stromal cells and their extracellular products. The TME serves an important role in tumor progression. Cancer-associated fibroblasts (CAFs) are the principal component of stromal cells within the TME, and contribute to tumor neo-angiogenesis by altering the proteome and degradome. The present paper reviews previous studies of the molecular signaling pathways by which CAFs promote tumor neo-angiogenesis and highlights therapeutic response targets. Also discussed are potential strategies for antitumor neo-angiogenesis to improve tumor treatment efficacy.
Collapse
Affiliation(s)
- Fang-Tao Wang
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Wei Sun
- Department of Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jing-Tao Zhang
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Yue-Zu Fan
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| |
Collapse
|
16
|
Kashima H, Noma K, Ohara T, Kato T, Katsura Y, Komoto S, Sato H, Katsube R, Ninomiya T, Tazawa H, Shirakawa Y, Fujiwara T. Cancer-associated fibroblasts (CAFs) promote the lymph node metastasis of esophageal squamous cell carcinoma. Int J Cancer 2018; 144:828-840. [PMID: 30367467 DOI: 10.1002/ijc.31953] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/29/2018] [Accepted: 10/11/2018] [Indexed: 12/11/2022]
Abstract
Lymph node metastasis is a pathognomonic feature of spreading tumors, and overcoming metastasis is a challenge in attaining more favorable clinical outcomes. Esophageal cancer is an aggressive tumor for which lymph node metastasis is a strong poor prognostic factor, and the tumor microenvironment (TME), and cancer-associated fibroblasts (CAFs) in particular, has been implicated in esophageal cancer progression. CAFs play a central role in the TME and have been reported to provide suitable conditions for the progression of esophageal cancer, similar to their role in other malignancies. However, little is known concerning the relevance of CAFs to the lymph node metastasis of esophageal cancer. Here, we used clinical samples of esophageal cancer to reveal that CAFs promote lymph node metastasis and subsequently verified the intercellular relationships in vitro and in vivo using an orthotopic metastatic mouse model. In the analysis of clinical samples, FAP+ CAFs were strongly associated with lymph node metastasis rather than with other prognostic factors. Furthermore, CAFs affected the ability of esophageal cancer cells to acquire metastatic phenotypes in vitro; this finding was confirmed by data from an in vivo orthotopic metastatic mouse model showing that the number of lymph node metastases increased upon injection of cocultured cancer cells and CAFs. In summary, we verified in vitro and in vivo that the accumulation of CAFs enhances the lymph node metastasis of ESCC. Our data suggest that CAF targeted therapy can reduce lymph node metastasis and improve the prognosis of patients with esophageal cancer in the future.
Collapse
Affiliation(s)
- Hajime Kashima
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiro Noma
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiaki Ohara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Pathology & Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuya Kato
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuki Katsura
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Komoto
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroaki Sato
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ryoichi Katsube
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takayuki Ninomiya
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Yasuhiro Shirakawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
17
|
Zhou J, Wang XH, Zhao YX, Chen C, Xu XY, Sun Q, Wu HY, Chen M, Sang JF, Su L, Tang XQ, Shi XB, Zhang Y, Yu Q, Yao YZ, Zhang WJ. Cancer-Associated Fibroblasts Correlate with Tumor-Associated Macrophages Infiltration and Lymphatic Metastasis in Triple Negative Breast Cancer Patients. J Cancer 2018; 9:4635-4641. [PMID: 30588247 PMCID: PMC6299377 DOI: 10.7150/jca.28583] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/22/2018] [Indexed: 12/25/2022] Open
Abstract
Background: Cancer-associated fibroblasts (CAFs) have been shown to be among the most prominent cells in tumor microenvironment and play a significant role in accelerating tumor metastasis by interacting with other type of cells. Tumor-associated macrophages (TAMs), the predominant tumor-infiltrating immune cells, also play important roles in cancer progression. Here, we aimed to evaluate the effects of CAFs on infiltration of TAMs and lymphatic metastasis in triple-negative breast cancer (TNBC). Material and methods: The study included 278 patients with histologically confirmed TNBC. Immunohistochemical staining of α-smooth muscle actin and fibroblast activation protein were used to identify CAFs. Polarized functional status of infiltrated TAMs was detected by expression of CD163. The clinicopathological features were assessed from all the patients' medical records. Results: The CAFs-related markers were found to be expressed more frequently in TNBC patents with aggressive behaviors, including recurrence and poor histological differentiation. High activation of CAFs was positively correlated with elevated infiltration of polarized CD163-positive TAMs and lymph node metastasis in TNBC patients. Multivariate Cox analysis revealed that the activation of CAFs, TAMs infiltration, and lymph node metastasis were independent prognostic factors for disease-free survival in TNBC patients. Conclusion: Cancer-associated fibroblasts were associated with infiltration of CD163-positive macrophages and lymphatic metastasis, and may be potential prognostic predictors of TNBC.
Collapse
Affiliation(s)
- Jing Zhou
- Dept. of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Dept. of General Surgery, Kunshan Fourth People's Hospital, Suzhou, China
| | - Xiao-Hua Wang
- Dept. of Medical Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | | | - Cheng Chen
- Dept. of Radiotherapy, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Xin-Yun Xu
- Dept. of Pathology, Affiliated Drum tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qi Sun
- Dept. of Pathology, Affiliated Drum tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Hong-Yan Wu
- Dept. of Pathology, Affiliated Drum tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Ming Chen
- Dept. of Pathology, Affiliated Drum tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jian-Feng Sang
- Dept. of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Lei Su
- Dept. of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiao-Qiao Tang
- Dept. of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xian-Biao Shi
- Dept. of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yin Zhang
- Dept. of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qiao Yu
- Dept. of Breast Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yong-Zhong Yao
- Dept. of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,School of Medicine, Southeast University
| | - Wei-Jie Zhang
- Dept. of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
18
|
Liu J, Huang C, Peng C, Xu F, Li Y, Yutaka Y, Xiong B, Yang X. Stromal fibroblast activation protein alpha promotes gastric cancer progression via epithelial-mesenchymal transition through Wnt/ β-catenin pathway. BMC Cancer 2018; 18:1099. [PMID: 30419872 PMCID: PMC6233532 DOI: 10.1186/s12885-018-5035-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 11/01/2018] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND To investigate the influence of fibroblast activation protein alpha (FAP) derived from cancer-associated fibroblasts (CAFs), as well as potential mechanism of epithelial mesenchymal transition (EMT), on gastric cancer (GC) progression. METHODS Correlation between CAFs-derived FAP and clinical results has been studied by using 60 GC cases. To confirm this relationship, SGC7901 cells were co-cultured with pre-established FAP-overexpressed fibroblasts in vitro and the characteristics including proliferation, migration, invasion and apoptosis abilities were detected subsequently. Meanwhile, SGC and GES1 cells cocultured with FAP-overexpressed fibroblasts were treated with cis-platinum for apoptotic analysis. The underlying EMT was detected by analyzing expression level of E-cadherin, ZO-1, N-cadherin, Vimentin, α-SMA, DKK1 and LEF-1 through western blot and immunofluorescence staining assay. Finally, the tumor-promoting ability of FAP was investigated by utlizing a xenograft gastric cancer nude mouse model. RESULTS It show that FAP has a high-risk correlation with the malignant level of clinical outcomes in GC patients. FAP promotes the ability of proliferation, migration, invasion, apoptosis-inhibition of SGC7901 cells and induces apoptosis of GES1 cells in vitro. The mechanism study shows that epithelial markers have been down-regulated and mesenchymal markers and Wnt/β-catenin signal pathway related proteins have been up-regulated. Animal assay suggests that tumor burden has been enhanced by FAP significantly in vivo. CONCLUSIONS Stromal FAP could be a potential prognostic biomarker in GC by promoting cancer progression via EMT through Wnt/ β-catenin signal pathway.
Collapse
Affiliation(s)
- Jiuyang Liu
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan, 430071, China.,Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chaoqun Huang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan, 430071, China
| | - Chunwei Peng
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan, 430071, China
| | - Fei Xu
- Department of General Surgery, Yingshan Renmin Hospital, Yingshan, 438700, China
| | - Yan Li
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Yonemura Yutaka
- Peritoneal Dissemination Center, Kishiwada Tokushukai Hospital, Kishiwada, 596-0032, Japan.,Department of Surgery, Kusatsu General Hospital, Shiga, 600-8189, Japan
| | - Bin Xiong
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan, 430071, China
| | - Xiaojun Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, China. .,Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan, 430071, China.
| |
Collapse
|
19
|
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: 230] [Impact Index Per Article: 32.9] [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
|
20
|
Avery D, Govindaraju P, Jacob M, Todd L, Monslow J, Puré E. Extracellular matrix directs phenotypic heterogeneity of activated fibroblasts. Matrix Biol 2018; 67:90-106. [PMID: 29248556 PMCID: PMC5910258 DOI: 10.1016/j.matbio.2017.12.003] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/08/2017] [Accepted: 12/10/2017] [Indexed: 12/17/2022]
Abstract
Activated fibroblasts are key players in the injury response, tumorigenesis, fibrosis, and inflammation. Dichotomous outcomes in response to varied stroma-targeted therapies in cancer emphasize the need to disentangle the roles of heterogeneous fibroblast subsets in physiological and pathophysiological settings. In wound healing, fibrosis, and myriad tumor types, fibroblast activation protein (FAP) and alpha-smooth muscle actin (αSMA) identify distinct, yet overlapping, activated fibroblast subsets. Prior studies established that FAPHi reactive fibroblasts and αSMAHi myofibroblasts can exert opposing influences in tumorigenesis. However, the factors that drive this phenotypic heterogeneity and the unique functional roles of these subsets have not been defined. We demonstrate that a convergence of ECM composition, elasticity, and transforming growth factor beta (TGF-β) signaling governs activated fibroblast phenotypic heterogeneity. Furthermore, FAPHi reactive fibroblasts and αSMAHi myofibroblasts exhibited distinct gene expression signatures and functionality in vitro, illuminating potentially unique roles of activated fibroblast subsets in tissue remodeling. These insights into activated fibroblast heterogeneity will inform the rational design of stroma-targeted therapies for cancer and fibrosis.
Collapse
Affiliation(s)
- Diana Avery
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, United States; Pharmacology Graduate Group of the University of Pennsylvania, Philadelphia, PA, United States
| | - Priya Govindaraju
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, United States; Pharmacology Graduate Group of the University of Pennsylvania, Philadelphia, PA, United States
| | - Michele Jacob
- Envision Pharma Group, Philadelphia, PA, United States
| | - Leslie Todd
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - James Monslow
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, United States; Pharmacology Graduate Group of the University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
21
|
Yazbeck R, Jaenisch SE, Abbott CA. Potential disease biomarkers: dipeptidyl peptidase 4 and fibroblast activation protein. PROTOPLASMA 2018; 255:375-386. [PMID: 28620698 DOI: 10.1007/s00709-017-1129-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/24/2017] [Indexed: 06/07/2023]
Abstract
The importance of the dipeptidyl peptidase 4 (DPP4) gene family in regulating critical biochemical pathways continues to emerge. The two most well-studied members of the family, DPP4 and fibroblast activation protein (FAP), have been investigated both as therapeutic targets for disease and as diagnostic biomarkers. The interest in DPP4 and FAP as potential disease biomarkers has been driven primarily by observations of altered expression profiles in inflammatory diseases and cancer. Furthermore, the stability and persistence of soluble DPP4 and FAP in the serum make them attractive candidate serology markers. This review summarises investigations into DPP4 and FAP as biomarkers of autoimmune disease, gut inflammation, psychosomatic disorders and malignancy and discusses their potential likelihood as clinically useful tools.
Collapse
Affiliation(s)
- Roger Yazbeck
- Department of Surgery, College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia, 5001, Australia
- Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, South Australia, Australia
| | - Simone E Jaenisch
- Department of Surgery, College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia, 5001, Australia
- Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, South Australia, Australia
| | - Catherine A Abbott
- Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, South Australia, Australia.
- College of Science and Engineering, Flinders University, GPO Box 2100, Adelaide, South Australia, 5001, Australia.
| |
Collapse
|
22
|
Hu M, Qian C, Hu Z, Fei B, Zhou H. Biomarkers in Tumor Microenvironment? Upregulation of Fibroblast Activation Protein-α Correlates with Gastric Cancer Progression and Poor Prognosis. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2017; 21:38-44. [PMID: 28206814 DOI: 10.1089/omi.2016.0159] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Gastric cancer is the third leading cause of cancer-related mortality worldwide. Recent evidence points to importance of cross talk between cancer cells and the surrounding stroma on gastric cancer progression. Tumor microenvironment biomarkers thus represent a new opportunity for diagnostics innovation. Reactive stromal fibroblasts selectively express the fibroblast activation protein alpha (FAP-α), a homodimeric integral membrane gelatinase that belongs to the serine protease family. We report here that FAP-α expression is significantly elevated in gastric cancer samples by more than fivefold (p < 0.05), using transcriptome data from The Cancer Genome Atlas. Notably, the greatest FAP-α upregulation was observed in the poorly differentiated group (p < 0.001). Moreover, elevated FAP-α expression levels correlated with adverse clinical-pathological characteristics, such as diffuse histological subtype (p < 0.001), advanced pathological stage (p < 0.01) and poor survival. Functional annotation analysis demonstrated that FAP-α upregulation was associated with activation of biological processes implicated in tumor progression, including cell migration and angiogenesis pathways. These observations underscore the possible prognostic significance of FAP-α in gastric cancer and its potential as a novel biomarker for personalized medicine. We caution, however, that further multiomics, biochemical, and animal studies are necessary to ascertain the role of FAP-α as a causative and mechanistic biomarker. Based on pathway analyses, we hypothesize that gastric cancer patients exhibiting FAP-α upregulation might presumably benefit from antiangiogenic drugs in addition to standard therapeutic regimens. We call for future research focusing on the tumor microenvironment biomarkers in clinical oncology.
Collapse
Affiliation(s)
- Mengmou Hu
- 1 Center of Laboratory Medicine, Danyang Hospital Affiliated to Nantong University , Danyang, 212300, China
| | - Chengjia Qian
- 2 Department of Gastrointestinal Surgery, Wuxi Fourth People's Hospital , Wuxi, China
| | - Ziwei Hu
- 3 Institute of Pharmaceutical Analysis, College of Pharmacy, Jinan University , Guangzhou, China
| | - Bojian Fei
- 2 Department of Gastrointestinal Surgery, Wuxi Fourth People's Hospital , Wuxi, China
| | - Haibo Zhou
- 3 Institute of Pharmaceutical Analysis, College of Pharmacy, Jinan University , Guangzhou, China
| |
Collapse
|
23
|
Koczorowska MM, Tholen S, Bucher F, Lutz L, Kizhakkedathu JN, De Wever O, Wellner UF, Biniossek ML, Stahl A, Lassmann S, Schilling O. Fibroblast activation protein-α, a stromal cell surface protease, shapes key features of cancer associated fibroblasts through proteome and degradome alterations. Mol Oncol 2015; 10:40-58. [PMID: 26304112 DOI: 10.1016/j.molonc.2015.08.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/24/2015] [Accepted: 08/03/2015] [Indexed: 12/23/2022] Open
Abstract
Cancer associated fibroblasts (CAFs) constitute an abundant stromal component of most solid tumors. Fibroblast activation protein (FAP) α is a cell surface protease that is expressed by CAFs. We corroborate this expression profile by immunohistochemical analysis of colorectal cancer specimens. To better understand the tumor-contextual role of FAPα, we investigate how FAPα shapes functional and proteomic features of CAFs using loss- and gain-of function cellular model systems. FAPα activity has a strong impact on the secreted CAF proteome ("secretome"), including reduced levels of anti-angiogenic factors, elevated levels of transforming growth factor (TGF) β, and an impact on matrix processing enzymes. Functionally, FAPα mildly induces sprout formation by human umbilical vein endothelial cells. Moreover, loss of FAPα leads to a more epithelial cellular phenotype and this effect was rescued by exogenous application of TGFβ. In collagen contraction assays, FAPα induced a more contractile cellular phenotype. To characterize the proteolytic profile of FAPα, we investigated its specificity with proteome-derived peptide libraries and corroborated its preference for cleavage carboxy-terminal to proline residues. By "terminal amine labeling of substrates" (TAILS) we explored FAPα-dependent cleavage events. Although FAPα acts predominantly as an amino-dipeptidase, putative FAPα cleavage sites in collagens are present throughout the entire protein length. In contrast, putative FAPα cleavage sites in non-collagenous proteins cluster at the amino-terminus. The degradomic study highlights cell-contextual proteolysis by FAPα with distinct positional profiles. Generally, our findings link FAPα to key aspects of CAF biology and attribute an important role in tumor-stroma interaction to FAPα.
Collapse
Affiliation(s)
- M M Koczorowska
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany
| | - S Tholen
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany
| | - F Bucher
- University Eye Hospital Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - L Lutz
- Institute of Surgical Pathology, Department of Pathology, University Medical Center, Freiburg, Germany
| | - J N Kizhakkedathu
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - O De Wever
- Laboratory of Experimental Cancer Research, Ghent University Hospital, 1P7, De Pintelaan 185, 9000 Gent, Belgium
| | - U F Wellner
- Clinic for Surgery, UKSH Campus Lübeck, Lübeck, Germany
| | - M L Biniossek
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany
| | - A Stahl
- University Eye Hospital Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - S Lassmann
- Institute of Surgical Pathology, Department of Pathology, University Medical Center, Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, D-79104 Freiburg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - O Schilling
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, D-79104 Freiburg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
24
|
Ammendola M, Sacco R, Marech I, Sammarco G, Zuccalà V, Luposella M, Patruno R, Giordano M, Ruggieri E, Zizzo N, Gadaleta CD, Ranieri G. Microvascular density and endothelial area correlate with Ki-67 proliferative index in surgically-treated pancreatic ductal adenocarcinoma patients. Oncol Lett 2015; 10:967-971. [PMID: 26622606 DOI: 10.3892/ol.2015.3286] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 04/09/2015] [Indexed: 12/14/2022] Open
Abstract
Previous experimental and clinical data have indicated that tumour cell proliferation is associated with angiogenesis; in addition, an increased microvascular density (MVD) of tumours has been associated with poor prognosis in solid and haematological malignancies. However, limited data exists regarding the association between tumour cell proliferation and angiogenesis in primary tumour tissue from pancreatic ductal adenocarcinoma (PDAC) patients; therefore, the present study aimed to investigate this association. A series of 31 PDAC patients with stage Tumour (T)2-3 Node (N)0-1 Metastasis (M)0 were recruited into the present study and subsequently underwent surgery. PDAC tissue and adjacent normal tissue (ANT), resected during surgery, were evaluated using immunohistochemistry and image analysis methods to determine MVD, endothelial area (EA) and Ki-67 expression, which is an indicator of cell proliferation rate. The results demonstrated a correlation between the above parameters with each other as well as the main clinico-pathological features of PDAC. Significant differences were identified in MVD, EA and Ki-67 proliferation index between PDAC and ANT. It was demonstrated that MVD, EA and Ki-67 proliferation index were significantly correlated with each other in tumour tissue (r=0.69-0.81; P=0.001-0.003). However, no other significant correlations were identified. These data therefore suggested that angiogenesis and cell proliferation rate were significantly increased in PDAC compared with ANT, which provides a biological basis for the potential use of novel combinations of angiogenesis inhibitors and anti-proliferative chemotherapeutic drugs in the treatment of PDAC.
Collapse
Affiliation(s)
- Michele Ammendola
- Department of Medical and Surgery Science, Clinical Surgery Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy ; Surgery Unit, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| | - Rosario Sacco
- Department of Medical and Surgery Science, Clinical Surgery Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy
| | - Ilaria Marech
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| | - Giuseppe Sammarco
- Department of Medical and Surgery Science, Clinical Surgery Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy
| | - Valeria Zuccalà
- Department of Health Science, Pathology Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy
| | - Maria Luposella
- Department of Medical and Surgery Science, Cardiovascular Disease Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy
| | - Rosa Patruno
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| | - Marcella Giordano
- Department of Medical and Surgery Science, Clinical Surgery Unit, University of Catanzaro 'Magna Graecia' Medical School, Catanzaro 88100, Italy
| | - Eustachio Ruggieri
- Surgery Unit, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| | - Nicola Zizzo
- Chair of Pathology, 'Aldo Moro' University of Bari, Bari 70010, Italy
| | - Cosmo Damiano Gadaleta
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| | - Girolamo Ranieri
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, 'Giovanni Paolo II', Bari 70124, Italy
| |
Collapse
|