1
|
Strell C, Rodríguez-Tomàs E, Östman A. Functional and clinical roles of stromal PDGF receptors in tumor biology. Cancer Metastasis Rev 2024; 43:1593-1609. [PMID: 38980580 PMCID: PMC11554757 DOI: 10.1007/s10555-024-10194-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024]
Abstract
PDGF receptors play pivotal roles in both developmental and physiological processes through the regulation of mesenchymal cells involved in paracrine instructive interactions with epithelial or endothelial cells. Tumor biology studies, alongside analyses of patient tissue samples, provide strong indications that the PDGF signaling pathways are also critical in various types of human cancer. This review summarizes experimental findings and correlative studies, which have explored the biological mechanisms and clinical relevance of PDGFRs in mesenchymal cells of the tumor microenvironment. Collectively, these studies support the overall concept that the PDGF system is a critical regulator of tumor growth, metastasis, and drug efficacy, suggesting yet unexploited targeting opportunities. The inter-patient variability in stromal PDGFR expression, as being linked to prognosis and treatment responses, not only indicates the need for stratified approaches in upcoming therapeutic investigations but also implies the potential for the development of PDGFRs as biomarkers of clinical utility, interestingly also in settings outside PDGFR-directed treatments.
Collapse
Affiliation(s)
- Carina Strell
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Bergen University, Bergen, Norway
| | | | - Arne Östman
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Bergen University, Bergen, Norway.
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
2
|
Zhou Q, Jin X, Zhao Y, Wang Y, Tao M, Cao Y, Yin X. Melanoma-associated fibroblasts in tumor-promotion flammation and antitumor immunity: novel mechanisms and potential immunotherapeutic strategies. Hum Mol Genet 2024; 33:1186-1193. [PMID: 38538564 PMCID: PMC11190611 DOI: 10.1093/hmg/ddae056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 06/22/2024] Open
Abstract
Melanoma, renowned for its aggressive behavior and resistance to conventional treatments, stands as a formidable challenge in the oncology landscape. The dynamic and complex interplay between cancer cells and the tumor microenvironment has gained significant attention, revealing Melanoma-Associated Fibroblasts (MAFs) as central players in disease progression. The heterogeneity of MAFs endows them with a dual role in melanoma. This exhaustive review seeks to not only shed light on the multifaceted roles of MAFs in orchestrating tumor-promoting inflammation but also to explore their involvement in antitumor immunity. By unraveling novel mechanisms underlying MAF functions, this review aims to provide a comprehensive understanding of their impact on melanoma development. Additionally, it delves into the potential of leveraging MAFs for innovative immunotherapeutic strategies, offering new avenues for enhancing treatment outcomes in the challenging realm of melanoma therapeutics.
Collapse
Affiliation(s)
- Qiujun Zhou
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Xiaoliang Jin
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Ying Zhao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Yueping Wang
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
- Department of First Clinical Medical College, Zhejiang Chinese Medical University, #548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310000, China
| | - Maocan Tao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
| | - Yi Cao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
| | - Xiaohu Yin
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), #54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
3
|
Chen Q, Luo J, Liu J, Yu H, Zhou M, Yu L, Chen Y, Zhang S, Mo Z. Integrating single-cell and spatial transcriptomics to elucidate the crosstalk between cancer-associated fibroblasts and cancer cells in hepatocellular carcinoma with spleen-deficiency syndrome. J Tradit Complement Med 2024; 14:321-334. [PMID: 38707923 PMCID: PMC11068993 DOI: 10.1016/j.jtcme.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/24/2023] [Accepted: 11/20/2023] [Indexed: 05/07/2024] Open
Abstract
Background and aim Most patients with hepatocellular carcinoma (HCC) in China have been diagnosed with spleen deficiency syndrome (SDS), which accelerates the progression of HCC by disrupting the tumor microenvironment homeostasis. This study aimed to investigate the intercellular crosstalk in HCC with SDS. Experimental procedure An HCC-SDS mouse model was established using orthotopic HCC transplantation based on reserpine-induced SDS. Single-cell data analysis and cancer cell prediction were conducted using Seurat and CopyKAT package, respectively. Intercellular interactions were explored using CellPhoneDB and CellChat and subsequently validated using co-culture assays, ELISA and histological staining. We performed pathway activity analysis using gene set variation analysis and the Seurat package. The extracellular matrix (ECM) remodeling was assessed using a gel contraction assay, atomic force microscopy, and Sirius red staining. The deconvolution of the spatial transcriptomics data using the "CARD" package based on single-cell data. Results and conclusion We successfully established the HCC-SDS mouse model. Twenty-nine clusters were identified. The interactions between cancer cells and cancer-associated fibroblasts (CAFs) were significantly enhanced via platelet-derived growth factor (PDGF) signaling in HCC-SDS. CAFs recruited in HCC-SDS lead to ECM remodeling and the activation of TGF-β signaling pathway. Deconvolution of the spatial transcriptome data revealed that CAFs physically surround cancer cells in HCC-SDS. This study reveals that the crosstalk of CAFs-cancer cells is crucial for the tumor-promoting effect of SDS. CAFs recruited by HCC via PDGFA may lead to ECM remodeling through activation of the TGF-β pathway, thereby forming a physical barrier to block immune cell infiltration under SDS.
Collapse
Affiliation(s)
- Qiuxia Chen
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Jin Luo
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Jiahui Liu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - He Yu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Meiling Zhou
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Ling Yu
- Department of Critical Care Medicine, The Second Clinical College to Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Yan Chen
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Shijun Zhang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | - Zhuomao Mo
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang Province, 311113, China
| |
Collapse
|
4
|
Sari D, Gozuacik D, Akkoc Y. Role of autophagy in cancer-associated fibroblast activation, signaling and metabolic reprograming. Front Cell Dev Biol 2024; 11:1274682. [PMID: 38234683 PMCID: PMC10791779 DOI: 10.3389/fcell.2023.1274682] [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: 08/08/2023] [Accepted: 12/08/2023] [Indexed: 01/19/2024] Open
Abstract
Tumors not only consist of cancerous cells, but they also harbor several normal-like cell types and non-cellular components. cancer-associated fibroblasts (CAFs) are one of these cellular components that are found predominantly in the tumor stroma. Autophagy is an intracellular degradation and quality control mechanism, and recent studies provided evidence that autophagy played a critical role in CAF formation, metabolic reprograming and tumor-stroma crosstalk. Therefore, shedding light on the autophagy and its role in CAF biology might help us better understand the roles of CAFs and the TME in cancer progression and may facilitate the exploitation of more efficient cancer diagnosis and treatment. Here, we provide an overview about the involvement of autophagy in CAF-related pathways, including transdifferentiation and activation of CAFs, and further discuss the implications of targeting tumor stroma as a treatment option.
Collapse
Affiliation(s)
- Dyana Sari
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Devrim Gozuacik
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
- Department of Medical Biology, School of Medicine, Koç University, Istanbul, Türkiye
- Department of Biotechnology, SUNUM Nanotechnology Research and Application Center, Istanbul, Türkiye
| | - Yunus Akkoc
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| |
Collapse
|
5
|
Zhang W, Wang J, Liu C, Li Y, Sun C, Wu J, Wu Q. Crosstalk and plasticity driving between cancer-associated fibroblasts and tumor microenvironment: significance of breast cancer metastasis. J Transl Med 2023; 21:827. [PMID: 37978384 PMCID: PMC10657029 DOI: 10.1186/s12967-023-04714-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are the most abundant stromal cell population in breast tumors. A functionally diverse population of CAFs increases the dynamic complexity of the tumor microenvironment (TME). The intertwined network of the TME facilitates the interaction between activated CAFs and breast cancer cells, which can lead to the proliferation and invasion of breast cells. Considering the special transmission function of CAFs, the aim of this review is to summarize and highlight the crosstalk between CAFs and breast cancer cells in the TME as well as the relationship between CAFs and extracellular matrix (ECM), soluble cytokines, and other stromal cells in the metastatic state. The crosstalk between cancer-associated fibroblasts and tumor microenvironment also provides a plastic therapeutic target for breast cancer metastasis. In the course of the study, the inhibitory effects of different natural compounds on targeting CAFs and the advantages of different drug combinations were summarized. CAFs are also widely used in the diagnosis and treatment of breast cancer. The cumulative research on this phenomenon supports the establishment of a targeted immune microenvironment as a possible breakthrough in the prevention of invasive metastasis of breast cancer.
Collapse
Affiliation(s)
- Wenfeng Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, China
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, 261000, China
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jia Wang
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, China
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, 261000, China
| | - Ye Li
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, China
| | - Changgang Sun
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, China.
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, 261000, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, China.
| |
Collapse
|
6
|
Gao D, Fang L, Liu C, Yang M, Yu X, Wang L, Zhang W, Sun C, Zhuang J. Microenvironmental regulation in tumor progression: Interactions between cancer-associated fibroblasts and immune cells. Biomed Pharmacother 2023; 167:115622. [PMID: 37783155 DOI: 10.1016/j.biopha.2023.115622] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/20/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023] Open
Abstract
The tumor microenvironment (TME), the "soil" on which tumor cells grow, has an important role in regulating the proliferation and metastasis of tumor cells as well as their response to treatment. Cancer-associated fibroblasts (CAFs), as the most abundant stromal cells of the TME, can not only directly alter the immunosuppressive effect of the TME through their own metabolism, but also influence the aggregation and function of immune cells by secreting a large number of cytokines and chemokines, reducing the body's immune surveillance of tumor cells and making them more prone to immune escape. Our study provides a comprehensive review of fibroblast chemotaxis, malignant transformation, metabolic characteristics, and interactions with immune cells. In addition, the current small molecule drugs targeting CAFs have been summarized, including both natural small molecules and targeted drugs for current clinical therapeutic applications. A complete review of the role of fibroblasts in TME from an immune perspective is presented, which has important implications in improving the efficiency of immunotherapy by targeting fibroblasts.
Collapse
Affiliation(s)
- Dandan Gao
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Liguang Fang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Mengrui Yang
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Xiaoyun Yu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Longyun Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China
| | - Wenfeng Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China; Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| |
Collapse
|
7
|
McCarthy GA, Di Niro R, Finan JM, Jain A, Guo Y, Wyatt C, Guimaraes A, Waugh T, Keith D, Morgan T, Sears R, Brody J. Deletion of the mRNA stability factor ELAVL1 (HuR) in pancreatic cancer cells disrupts the tumor microenvironment integrity. NAR Cancer 2023; 5:zcad016. [PMID: 37089813 PMCID: PMC10113877 DOI: 10.1093/narcan/zcad016] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/08/2023] [Accepted: 04/06/2023] [Indexed: 04/25/2023] Open
Abstract
Stromal cells promote extensive fibrosis in pancreatic ductal adenocarcinoma (PDAC), which is associated with poor prognosis and therapeutic resistance. We report here for the first time that loss of the RNA-binding protein human antigen R (HuR, ELAVL1) in PDAC cells leads to reprogramming of the tumor microenvironment. In multiple in vivo models, CRISPR deletion of ELAVL1 in PDAC cells resulted in a decrease of collagen deposition, accompanied by a decrease of stromal markers (i.e. podoplanin, α-smooth muscle actin, desmin). RNA-sequencing data showed that HuR plays a role in cell-cell communication. Accordingly, cytokine arrays identified that HuR regulates the secretion of signaling molecules involved in stromal activation and extracellular matrix organization [i.e. platelet-derived growth factor AA (PDGFAA) and pentraxin 3]. Ribonucleoprotein immunoprecipitation analysis and transcription inhibition studies validated PDGFA mRNA as a novel HuR target. These data suggest that tumor-intrinsic HuR supports extrinsic activation of the stroma to produce collagen and desmoplasia through regulating signaling molecules (e.g. PDGFAA). HuR-deficient PDAC in vivo tumors with an altered tumor microenvironment are more sensitive to the standard of care gemcitabine, as compared to HuR-proficient tumors. Taken together, we identified a novel role of tumor-intrinsic HuR in its ability to modify the surrounding tumor microenvironment and regulate PDGFAA.
Collapse
Affiliation(s)
- Grace A McCarthy
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Roberto Di Niro
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Jennifer M Finan
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Aditi Jain
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Yifei Guo
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Cory R Wyatt
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, OR 97239, USA
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alexander R Guimaraes
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, OR 97239, USA
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Trent A Waugh
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR 97201, USA
| | - Dove Keith
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR 97201, USA
| | - Terry K Morgan
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rosalie C Sears
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR 97201, USA
| | - Jonathan R Brody
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| |
Collapse
|
8
|
Lee YE, Go GY, Koh EY, Yoon HN, Seo M, Hong SM, Jeong JH, Kim JC, Cho D, Kim TS, Kim SC, Jun E, Jang M. Synergistic therapeutic combination with a CAF inhibitor enhances CAR-NK-mediated cytotoxicity via reduction of CAF-released IL-6. J Immunother Cancer 2023; 11:jitc-2022-006130. [PMID: 36849201 PMCID: PMC9972461 DOI: 10.1136/jitc-2022-006130] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) in the tumor microenvironment (TME) contribute to an impaired functionality of natural killer (NK) cells that have emerged as a promising therapeutic modality. The interaction between CAFs and NK cells within the TME exerts major inhibitory effects on immune responses, indicating CAF-targeted therapies as potential targets for effective NK-mediated cancer killing. METHODS To overcome CAF-induced NK dysfunction, we selected an antifibrotic drug, nintedanib, for synergistic therapeutic combination. To evaluate synergistic therapeutic efficacy, we established an in vitro 3D Capan2/patient-derived CAF spheroid model or in vivo mixed Capan2/CAF tumor xenograft model. The molecular mechanism of NK-mediated synergistic therapeutic combination with nintedanib was revealed through in vitro experiments. In vivo therapeutic combination efficacy was subsequently evaluated. Additionally, the expression score of target proteins was measured in patient-derived tumor sections by the immunohistochemical method. RESULTS Nintedanib blocked the platelet-derived growth factor receptor β (PDGFRβ) signaling pathway and diminished the activation and growth of CAFs, markedly reducing CAF-secreted IL-6. Moreover, coadministration of nintedanib improved the mesothelin (MSLN) targeting chimeric antigen receptor-NK-mediated tumor killing abilities in CAF/tumor spheroids or a xenograft model. The synergistic combination resulted in intense NK infiltration in vivo. Nintedanib alone exerted no effects, whereas blockade of IL-6 trans-signaling ameliorated the function of NK cells. The combination of the expression of MSLN and the PDGFRβ+-CAF population area, a potential prognostic/therapeutic marker, was associated with inferior clinical outcomes. CONCLUSION Our strategy against PDGFRβ+-CAF-containing pancreatic cancer allows improvements in the therapy of pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Young Eun Lee
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Korea (the Republic of),Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea (the Republic of)
| | - Ga-Yeon Go
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Korea (the Republic of)
| | - Eun-Young Koh
- Department of Convergence Medicine, Asan Institute for Life Sciences, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea (the Republic of)
| | - Han-Na Yoon
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Korea (the Republic of)
| | - Minkoo Seo
- Corporate Research & Development Center, UCI therapeutics, Seoul, Korea (the Republic of)
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea (the Republic of)
| | - Ji Hye Jeong
- Department of Convergence Medicine, Asan Institute for Life Sciences, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea (the Republic of)
| | - Jin-Chul Kim
- Natural Product Research Center, Institute of Natural Products, Korea Institute of Science and Technology, Gangneung, Korea (the Republic of)
| | - Duck Cho
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea (the Republic of),Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea (the Republic of)
| | - Tae Sung Kim
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea (the Republic of)
| | - Song Cheol Kim
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea (the Republic of)
| | - Eunsung Jun
- Department of Convergence Medicine, Asan Institute for Life Sciences, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea (the Republic of) .,Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea (the Republic of)
| | - Mihue Jang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Korea (the Republic of) .,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Korea (the Republic of)
| |
Collapse
|
9
|
Suppression of Platelet-Derived Growth Factor Receptor-Alpha Overcomes Resistance to Trastuzumab through STAT3-Dependent IL-6 Reduction in HER2-Positive Breast Cancer Cells. Biomedicines 2023; 11:biomedicines11030675. [PMID: 36979654 PMCID: PMC10045855 DOI: 10.3390/biomedicines11030675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 02/25/2023] Open
Abstract
Platelet-derived growth factor receptor (PDGFR) plays an essential role in the proliferation and invasion of malignant cancer cells. However, the functional role of PDGFR alpha (PDGFRA) in HER2-positive (HER2+) breast cancer has not been fully clarified yet. Thus, the objective of this study was to investigate the clinical significance of PDGFRA and the therapeutic potential of PDGFR inhibitors as part of an effort to overcome trastuzumab (TRZ) resistance. Aberrant PDGFRA expression is closely associated with decreased survival in HER2+ breast cancers. Therefore, we established BT474 trastuzumab-sensitive (TRZ_S) and trastuzumab-resistant (TRZ_R) cells to investigate the association between PDGFR signaling and TRZ resistance. We found that PDGFRA was significantly upregulated in the BT474 TRZ_R cells. In addition, IL-6 expression, which was also found to be upregulated in the TRZ_R cells, was induced by PDGFC, a ligand of PDGFR. Next, we investigated the effects of ponatinib and sunitinib, PDGFR inhibitors, on the BT474 TRZ_R and HCC1954 (TRZ-resistant cell line) cells. These inhibitors decreased cell viability and migration in a dose-dependent manner. Additionally, IL-6 expression was decreased by ponatinib in both the BT474 TRZ_R and HCC1954 cells. In contrast, IL-6 was not suppressed by TRZ, implying that the PDGFRA/STAT3/IL-6 axis is associated with resistance to TRZ. In addition, we found that STAT3 and ERK phosphorylation were increased in the BT474 TRZ_R cells. IL-6 expression was suppressed by a STAT3 inhibitor, indicating that IL-6 expression is modulated downstream of STAT3. Taken together, these results suggest that PDGFRA could serve as a therapeutic target to overcome TRZ resistance.
Collapse
|
10
|
Toledo B, Picon-Ruiz M, Marchal JA, Perán M. Dual Role of Fibroblasts Educated by Tumour in Cancer Behavior and Therapeutic Perspectives. Int J Mol Sci 2022; 23:15576. [PMID: 36555218 PMCID: PMC9778751 DOI: 10.3390/ijms232415576] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Tumours are complex systems with dynamic interactions between tumour cells, non-tumour cells, and extracellular components that comprise the tumour microenvironment (TME). The majority of TME's cells are cancer-associated fibroblasts (CAFs), which are crucial in extracellular matrix (ECM) construction, tumour metabolism, immunology, adaptive chemoresistance, and tumour cell motility. CAF subtypes have been identified based on the expression of protein markers. CAFs may act as promoters or suppressors in tumour cells depending on a variety of factors, including cancer stage. Indeed, CAFs have been shown to promote tumour growth, survival and spread, and secretome changes, but they can also slow tumourigenesis at an early stage through mechanisms that are still poorly understood. Stromal-cancer interactions are governed by a variety of soluble factors that determine the outcome of the tumourigenic process. Cancer cells release factors that enhance the ability of fibroblasts to secrete multiple tumour-promoting chemokines, acting on malignant cells to promote proliferation, migration, and invasion. This crosstalk between CAFs and tumour cells has given new prominence to the stromal cells, from being considered as mere physical support to becoming key players in the tumour process. Here, we focus on the concept of cancer as a non-healing wound and the relevance of chronic inflammation to tumour initiation. In addition, we review CAFs heterogeneous origins and markers together with the potential therapeutic implications of CAFs "re-education" and/or targeting tumour progression inhibition.
Collapse
Affiliation(s)
- Belén Toledo
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
| | - Manuel Picon-Ruiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| |
Collapse
|
11
|
Xue XM, Liu YY, Chen XM, Tao BY, Liu P, Zhou HW, Zhang C, Wang L, Jiang YK, Ding ZW, Shen WD, Zhang J, Yang SM, Wang FY. Pan-cancer analysis identifies NT5E as a novel prognostic biomarker on cancer-associated fibroblasts associated with unique tumor microenvironment. Front Pharmacol 2022; 13:1064032. [PMID: 36569293 PMCID: PMC9768042 DOI: 10.3389/fphar.2022.1064032] [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: 10/07/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Ecto-5'-nucleotidase (NT5E) encodes the cluster of differentiation 73 (CD73), whose overexpression contributes to the formation of immunosuppressive tumor microenvironment and is related to exacerbated prognosis, increased risk of metastasis and resistance to immunotherapy of various tumors. However, the prognostic significance of NT5E in pan-cancer is obscure so far. Methods: We explored the expression level of NT5E in cancers and adjacent tissues and revealed the relationship between the NT5E expression level and clinical outcomes in pan-cancer by utilizing the UCSC Xena database. Then, correlation analyses were performed to evaluate the relationship between NT5E expression and immune infiltration level via EPIC, MCP-counter and CIBERSORT methods, and the enrichment analysis were employed to identify NT5E-interacting molecules and functional pathways. Furthermore, we conducted single-cell analysis to explore the potential role of NT5E on single-cell level based on the CancerSEA database. Meanwhile, gene set enrichment analysis (GSEA) in single-cell level was also conducted in TISCH database and single-cell signature explorer was utilized to evaluate the epithelial-mesenchymal transition (EMT) level in each cell type. Results: The expression level of NT5E was aberrant in almost all cancer types, and was correlated with worse prognosis in several cancers. Notably, NT5E overexpression was related to worse overall survival (OS) in pancreatic adenocarcinoma (PAAD), head and neck squamous cell carcinoma (HNSC), mesothelioma (MESO), stomach adenocarcinoma (STAD), uveal melanoma (UVM) and cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) (p < 0.01). NT5E-related immune microenvironment analysis revealed that NT5E is associated positively with the degree of infiltration of cancer-associated fibroblasts (CAFs) and endothelial cells in most cancers. Enrichment analysis of cellular component (CC) demonstrated the critical part of NT5E played in cell-substrate junction, cell-substrate adherens junction, focal adhesion and external side of plasma membrane. Finally, single-cell analysis of NT5E illuminated that EMT function of CAFs was elevated in basal cell carcinoma (BCC), skin cutaneous melanoma (SKCM), HNSC and PAAD. Conclusion: NT5E could serve as a potential prognostic biomarker for cancers. The potential mechanism may be related to the upregulated EMT function of CAFs, which provides novel inspiration for immunotherapy by targeting CAFs with high NT5E expression.
Collapse
Affiliation(s)
- Xin-miao Xue
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Yu-yang Liu
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Department of Neurosurgery, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xue-min Chen
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Bing-yan Tao
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Department of Neurosurgery, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Peng Liu
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Han-wen Zhou
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Chi Zhang
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,The Zhantansi Outpatient Department of Central Medical Branch of People’s Liberation Army (PLA) General Hospital Beijing, China
| | - Li Wang
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Yu-ke Jiang
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Zhi-wei Ding
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China,Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Wei-dong Shen
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China
| | - Jun Zhang
- Department of Neurosurgery, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China,*Correspondence: Jun Zhang, ; Shi-ming Yang, ; Fang-yuan Wang,
| | - Shi-ming Yang
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China,*Correspondence: Jun Zhang, ; Shi-ming Yang, ; Fang-yuan Wang,
| | - Fang-yuan Wang
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese People’s Liberation Army (PLA) General Hospital, National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Beijing Key Lab of Hearing Impairment Prevention and Treatment, Ministry of Education, Beijing, China,*Correspondence: Jun Zhang, ; Shi-ming Yang, ; Fang-yuan Wang,
| |
Collapse
|
12
|
Diverse roles of tumor-stromal PDGFB-to-PDGFRβ signaling in breast cancer growth and metastasis. Adv Cancer Res 2022; 154:93-140. [PMID: 35459473 DOI: 10.1016/bs.acr.2022.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Over the last couple of decades, it has become increasingly apparent that the tumor microenvironment (TME) mediates every step of cancer progression and solid tumors are only able to metastasize with a permissive TME. This intricate interaction of cancer cells with their surrounding TME, or stroma, is becoming more understood with an ever greater knowledge of tumor-stromal signaling pairs such as platelet-derived growth factors (PDGF) and their cognate receptors. We and others have focused our research efforts on understanding how tumor-derived PDGFB activates platelet-derived growth factor receptor beta (PDGFRβ) signaling specifically in the breast cancer TME. In this chapter, we broadly discuss PDGF and PDGFR expression patterns and signaling in normal physiology and breast cancer. We then detail the expansive roles played by the PDGFB-to-PDGFRβ signaling pathway in modulating breast tumor growth and metastasis with a focus on specific cellular populations within the TME, which are responsive to tumor-derived PDGFB. Given the increasingly appreciated importance of PDGFB-to-PDGFRβ signaling in breast cancer progression, specifically in promoting metastasis, we end by discussing how therapeutic targeting of PDGFB-to-PDGFRβ signaling holds great promise for improving current breast cancer treatment strategies.
Collapse
|
13
|
Yang HH, Liu JW, Lee JH, Harn HJ, Chiou TW. Pancreatic Adenocarcinoma Therapeutics Targeting RTK and TGF Beta Receptor. Int J Mol Sci 2021; 22:ijms22158125. [PMID: 34360896 PMCID: PMC8348294 DOI: 10.3390/ijms22158125] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/28/2021] [Accepted: 07/26/2021] [Indexed: 12/21/2022] Open
Abstract
Despite the improved overall survival rates in most cancers, pancreatic cancer remains one of the deadliest cancers in this decade. The rigid microenvironment, which majorly comprises cancer-associated fibroblasts (CAFs), plays an important role in the obstruction of pancreatic cancer therapy. To overcome this predicament, the signaling of receptor tyrosine kinases (RTKs) and TGF beta receptor (TGFβR) in both pancreatic cancer cell and supporting CAF should be considered as the therapeutic target. The activation of receptors has been reported to be aberrant to cell cycle regulation, and signal transduction pathways, such as growth-factor induced proliferation, and can also influence the apoptotic sensitivity of tumor cells. In this article, the regulation of RTKs/TGFβR between pancreatic ductal adenocarcinoma (PDAC) and CAFs, as well as the RTKs/TGFβR inhibitor-based clinical trials on pancreatic cancer are reviewed.
Collapse
Affiliation(s)
- Hsin-Han Yang
- Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien 974, Taiwan;
| | - Jen-Wei Liu
- Everfront Biotech Inc., New Taipei City 221, Taiwan; (J.-W.L.); (J.-H.L.)
| | - Jui-Hao Lee
- Everfront Biotech Inc., New Taipei City 221, Taiwan; (J.-W.L.); (J.-H.L.)
| | - Horng-Jyh Harn
- Bioinnovation Center, Tzu Chi Foundation, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien 970, Taiwan
- Department of Pathology, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien 970, Taiwan
- Correspondence: (H.-J.H.); (T.-W.C.)
| | - Tzyy-Wen Chiou
- Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien 974, Taiwan;
- Correspondence: (H.-J.H.); (T.-W.C.)
| |
Collapse
|
14
|
Receptor tyrosine kinases and cancer: oncogenic mechanisms and therapeutic approaches. Oncogene 2021; 40:4079-4093. [PMID: 34079087 DOI: 10.1038/s41388-021-01841-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/29/2021] [Accepted: 05/13/2021] [Indexed: 02/05/2023]
Abstract
Receptor tyrosine kinases (RTKs) are transmembrane receptors of great clinical interest due to their role in disease, notably cancer. Since their discovery, several mechanisms of RTK dysregulation have been identified, resulting in multiple cancer types displaying 'oncogenic addiction' to RTKs. As a result, RTKs have represented a major class for targeted therapeutics over the past two decades, with numerous small molecule-based tyrosine kinase inhibitor (TKI) therapeutics having been developed and clinically approved for several cancers. However, many of the current RTK inhibitor treatments eventually result in the rapid development of acquired resistance and subsequent tumor relapse. Recent technological advances and tools are being generated for the identification of novel RTK small molecule therapeutics. These newer technologies will be important for the identification of diverse types of RTK inhibitors, targeting both the receptors themselves as well as key cellular factors that play important roles in the RTK signaling cascade.
Collapse
|
15
|
The effect of platelet G proteins on platelet extravasation and tumor growth in the murine model of ovarian cancer. Blood Adv 2021; 5:1947-1951. [PMID: 33821990 DOI: 10.1182/bloodadvances.2020003410] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/02/2021] [Indexed: 12/21/2022] Open
Abstract
We and other investigators have shown that platelets promote metastasis and the growth of tumors. Our rationale for conducting this study is that platelets' prometastatic and progrowth effects depend on a close encounter between platelets and cancer cells. This interaction occurs inside blood vessels with circulating tumor cells and outside blood vessels with cancer cells residing in the tumor parenchyma. Our hypothesis was that platelet extravasation is required for the effect of platelets on tumor growth. Platelets respond to environmental stimuli by activation of G protein-coupled receptors on their surface. We investigated the impact of various platelet G proteins on the growth of ovarian cancer tumors and platelet extravasation. We used mice with platelet-specific deficiency of Gαi2 (Gi), Gα13 (G13), or Gαq (Gq) in a syngeneic ovarian cancer model. We measured the total weight of tumor nodules resected from tumor-bearing mice. We developed methods for automated whole-slide image acquisition and unbiased computerized image analysis to quantify extravasated platelets. We compared the number of platelets inside tumor nodules of platelet G protein-deficient tumor-bearing mice. We found that deficiency of Gi and G13, but not Gq, in platelets resulted in smaller tumors compared with those in corresponding littermates. Deficiency of Gi and G13 in platelets reduced the number of extravasated platelets by >90%, but deficiency of Gq did not reduce the number of extravasated platelets significantly. The lack of Gi or G13 in platelets reduced platelet extravasation into the tumor and tumor growth.
Collapse
|
16
|
In-Depth Characterization of Stromal Cells within the Tumor Microenvironment Yields Novel Therapeutic Targets. Cancers (Basel) 2021; 13:cancers13061466. [PMID: 33806802 PMCID: PMC8005121 DOI: 10.3390/cancers13061466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/23/2022] Open
Abstract
Simple Summary This up-to-date and in-depth review describes fibroblast-derived cells and their role within the tumor microenvironment for tumor progression. Moreover, targets for future antitumor therapies are summarized and potential aspects for future translational research are outlined. Furthermore, this review discusses the challenges and possible obstacles related to certain treatment targets. Abstract Cells within the tumor stroma are essential for tumor progression. In particular, cancer-associated fibroblasts (CAF) and CAF precursor cells (resident fibroblasts and mesenchymal stromal cells) are responsible for the formation of the extracellular matrix in tumor tissue. Consequently, CAFs directly and indirectly mediate inflammation, metastasis, immunomodulation, angiogenesis, and the development of tumor chemoresistance, which is orchestrated by complex intercellular cytokine-mediated crosstalk. CAFs represent a strategic target in antitumor therapy but their heterogeneity hinders effective treatment regimes. In-depth understanding of CAF subpopulations and knowledge of specific functions in tumor progression will ultimately result in more specific and effective cancer treatments. This review provides a detailed description of CAFs and CAF precursor cells and summarizes possible treatment strategies as well as molecular targets of these cells in antitumor therapies.
Collapse
|
17
|
In Vitro Suppression of T Cell Proliferation Is a Conserved Function of Primary and Immortalized Human Cancer-Associated Fibroblasts. Int J Mol Sci 2021; 22:ijms22041827. [PMID: 33673197 PMCID: PMC7918788 DOI: 10.3390/ijms22041827] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
T cell immunotherapy is now a mainstay therapy for several blood-borne cancers as well as metastatic melanoma. Unfortunately, many epithelial tumors respond poorly to immunotherapy, and the reasons for this are not well understood. Cancer-associated fibroblasts (CAFs) are the most frequent non-neoplastic cell type in most solid tumors, and they are emerging as a key player in immunotherapy resistance. A range of immortalized CAF lines will be essential tools that will allow us to understand immune responses against cancer and develop novel strategies for cancer immunotherapy. To study the effect of CAFs on T cell proliferation, we created and characterized a number of novel immortalized human CAFs lines (Im-CAFs) from human breast, colon, and pancreatic carcinomas. Im-CAFs shared similar phenotypes, matrix remodeling and contraction capabilities, and growth and migration rates compared to the primary CAFs. Using primary isolates from breast carcinoma, colorectal carcinoma, and pancreatic ductal adenocarcinoma, we report that CAFs across major tumor types are able to potently suppress T cell proliferation in vitro. Im-CAFs retained this property. Im-CAFs are a key tool that will provide important insights into the mechanisms of CAF-mediated T cell suppression through techniques such as CRISPR-Cas9 modification, molecular screens, and pipeline drug testing.
Collapse
|
18
|
Louault K, Li RR, DeClerck YA. Cancer-Associated Fibroblasts: Understanding Their Heterogeneity. Cancers (Basel) 2020; 12:cancers12113108. [PMID: 33114328 PMCID: PMC7690906 DOI: 10.3390/cancers12113108] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/12/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Cancer-associated fibroblasts (CAFs) play an important contributory role in the microenvironment of tumors. They originate from different cells, have multiple pro-and anti-tumorigenic functions in tumors and their presence is variable among cancer types. Recently, there has been evidence that CAFs represent a highly heterogeneous group of cells that can now be characterized and identified at the single cell level. This review article summarizes our recent understanding of the highly heterogeneous nature of the origin, phenotype and function of CAFs and how such understanding will lead to a more precise approach to target or use CAFs and their precursor cells in the treatment of cancer. Abstract The tumor microenvironment (TME) plays a critical role in tumor progression. Among its multiple components are cancer-associated fibroblasts (CAFs) that are the main suppliers of extracellular matrix molecules and important contributors to inflammation. As a source of growth factors, cytokines, chemokines and other regulatory molecules, they participate in cancer progression, metastasis, angiogenesis, immune cell reprogramming and therapeutic resistance. Nevertheless, their role is not fully understood, and is sometimes controversial due to their heterogeneity. CAFs are heterogeneous in their origin, phenotype, function and presence within tumors. As a result, strategies to target CAFs in cancer therapy have been hampered by the difficulties in better defining the various populations of CAFs and by the lack of clear recognition of their specific function in cancer progression. This review discusses how a greater understanding of the heterogeneous nature of CAFs could lead to better approaches aimed at their use or at their targeting in the treatment of cancer.
Collapse
Affiliation(s)
- Kévin Louault
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90027, USA
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
- Correspondence: or (K.L); (Y.A.D.); Tel.: +1-323-361-5649 (Y.A.D.); Fax: +1-323-361-4902 (Y.A.D.)
| | - Rong-Rong Li
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA;
| | - Yves A. DeClerck
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90027, USA
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Biochemistry and Molecular Biology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
- Correspondence: or (K.L); (Y.A.D.); Tel.: +1-323-361-5649 (Y.A.D.); Fax: +1-323-361-4902 (Y.A.D.)
| |
Collapse
|
19
|
Sözmen M, Devrim AK, Sudağıdan M, Kabak YB, Yıldırım F. Expression of angiogenic growth factors in canine squamous cell cancers. Biotech Histochem 2020; 96:450-459. [PMID: 33006294 DOI: 10.1080/10520295.2020.1818826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Skin and subcutaneous tissue tumors are the most common neoplasms in dogs. The most common sites of origin in dogs include digits, skin and the oral cavity including cheek and retromandibular area. We investigated canine squamous cell carcinoma (SCC) samples from 15 dogs and classified them histopathologically according to the degree of differentiation. bFGF, VEGF-C, TGF-β, PDGF-A, PDGF-C and PDGFR-α expressions were assessed using immunohistochemistry to determine the roles of growth factors during SCC. We found that TGF-β1 immunolabeling was elevated significantly compared to healthy controls in SCC originating from nailbeds, while expression of other growth factors did not change significantly. Our findings might explain the role of TGF-β1 in the infiltrative and malignant behavior of SCC originating from nailbeds.
Collapse
Affiliation(s)
- M Sözmen
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - A K Devrim
- Department of Biochemistry, Faculty of Veterinary Medicine, Kirikkale University, Kirikkale, Turkey
| | - M Sudağıdan
- KIT-ARGEM R & D Center, Konya Food and Agriculture University, Konya, Turkey
| | - Y B Kabak
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - F Yıldırım
- Department of Pathology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
20
|
Lai YT, Chao HW, Lai ACY, Lin SH, Chang YJ, Huang YS. CPEB2-activated PDGFRα mRNA translation contributes to myofibroblast proliferation and pulmonary alveologenesis. J Biomed Sci 2020; 27:52. [PMID: 32295602 PMCID: PMC7160907 DOI: 10.1186/s12929-020-00643-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 03/26/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alveologenesis is the final stage of lung development to form air-exchanging units between alveoli and blood vessels. Genetic susceptibility or hyperoxic stress to perturb this complicated process can cause abnormal enlargement of alveoli and lead to bronchopulmonary dysplasia (BPD)-associated emphysema. Platelet-derived growth factor receptor α (PDGFRα) signaling is crucial for alveolar myofibroblast (MYF) proliferation and its deficiency is associated with risk of BPD, but posttranscriptional mechanisms regulating PDGFRα synthesis during lung development remain largely unexplored. Cytoplasmic polyadenylation element-binding protein 2 (CPEB2) is a sequence-specific RNA-binding protein and translational regulator. Because CPEB2-knockout (KO) mice showed emphysematous phenotypes, we investigated how CPEB2-controlled translation affects pulmonary development and function. METHODS Respiratory and pulmonary functions were measured by whole-body and invasive plethysmography. Histological staining and immunohistochemistry were used to analyze morphology, proliferation, apoptosis and cell densities from postnatal to adult lungs. Western blotting, RNA-immunoprecipitation, reporter assay, primary MYF culture and ectopic expression rescue were performed to demonstrate the role of CPEB2 in PDGFRα mRNA translation and MYF proliferation. RESULTS Adult CPEB2-KO mice showed emphysema-like dysfunction. The alveolar structure in CPEB2-deficient lungs appeared normal at birth but became simplified through the alveolar stage of lung development. In CPEB2-null mice, we found reduced proliferation of MYF progenitors during alveolarization, abnormal deposition of elastin and failure of alveolar septum formation, thereby leading to enlarged pulmonary alveoli. We identified that CPEB2 promoted PDGFRα mRNA translation in MYF progenitors and this positive regulation could be disrupted by H2O2, a hyperoxia-mimetic treatment. Moreover, decreased proliferating ability in KO MYFs due to insufficient PDGFRα expression was rescued by ectopic expression of CPEB2, suggesting an important role of CPEB2 in upregulating PDGFRα signaling for pulmonary alveologenesis. CONCLUSIONS CPEB2-controlled translation, in part through promoting PDGFRα expression, is indispensable for lung development and function. Since defective pulmonary PDGFR signaling is a key feature of human BPD, CPEB2 may be a risk factor for BPD.
Collapse
Affiliation(s)
- Yen-Ting Lai
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd, Taipei, 11529, Taiwan
| | - Hsu-Wen Chao
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Alan Chuan-Ying Lai
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd, Taipei, 11529, Taiwan
| | - Shu-Hui Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ya-Jen Chang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd, Taipei, 11529, Taiwan.
| | - Yi-Shuian Huang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd, Taipei, 11529, Taiwan.
| |
Collapse
|
21
|
Nurmik M, Ullmann P, Rodriguez F, Haan S, Letellier E. In search of definitions: Cancer-associated fibroblasts and their markers. Int J Cancer 2020; 146:895-905. [PMID: 30734283 PMCID: PMC6972582 DOI: 10.1002/ijc.32193] [Citation(s) in RCA: 404] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/14/2018] [Accepted: 01/28/2019] [Indexed: 12/12/2022]
Abstract
The tumor microenvironment has been identified as one of the driving factors of tumor progression and invasion. Inside this microenvironment, cancer-associated fibroblasts (CAFs), a type of perpetually activated fibroblasts, have been implicated to have a strong tumor-modulating effect and play a key role in areas such as drug resistance. Identification of CAFs has typically been carried based on the expression of various "CAF markers", such as fibroblast activation protein alpha (FAP) and alpha smooth muscle actin (αSMA), which separates them from the larger pool of fibroblasts present in the body. However, as outlined in this Review, the expression of various commonly used fibroblast markers is extremely heterogeneous and varies strongly between different CAF subpopulations. As such, novel selection methods based on cellular function, as well as further characterizing research, are vital for the standardization of CAF identification in order to improve the cross-applicability of different research studies in the field. The aim of this review is to give a thorough overview of the commonly used fibroblast markers in the field and their various strengths and, more importantly, their weaknesses, as well as to highlight potential future avenues for CAF identification and targeting.
Collapse
Affiliation(s)
- Martin Nurmik
- Molecular Disease Mechanisms Group, Life Sciences Research UnitUniversity of LuxembourgBelvauxLuxembourg
| | - Pit Ullmann
- Molecular Disease Mechanisms Group, Life Sciences Research UnitUniversity of LuxembourgBelvauxLuxembourg
| | - Fabien Rodriguez
- Molecular Disease Mechanisms Group, Life Sciences Research UnitUniversity of LuxembourgBelvauxLuxembourg
| | - Serge Haan
- Molecular Disease Mechanisms Group, Life Sciences Research UnitUniversity of LuxembourgBelvauxLuxembourg
| | - Elisabeth Letellier
- Molecular Disease Mechanisms Group, Life Sciences Research UnitUniversity of LuxembourgBelvauxLuxembourg
| |
Collapse
|
22
|
Tang Y, Lu Y, Chen Y, Luo L, Cai L, Peng B, Huang W, Liao H, Zhao L, Pan M. Pre-metastatic niche triggers SDF-1/CXCR4 axis and promotes organ colonisation by hepatocellular circulating tumour cells via downregulation of Prrx1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:473. [PMID: 31752959 PMCID: PMC6873584 DOI: 10.1186/s13046-019-1475-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 11/07/2019] [Indexed: 01/03/2023]
Abstract
Background Circulating tumour cells (CTCs), especially mesenchymal CTCs, are important determinants of metastasis, which leads to most recurrence and mortality in hepatocellular carcinoma (HCC). However, little is known about the underlying mechanisms of CTC colonisation in pre-metastatic niches. Methods Detection and classification of CTCs in patients were performed using the CanPatrol™ system. A lentiviral vector expressing Prrx1-targeting shRNA was constructed to generate a stable HCC cell line with low expression of Prrx1. The effect of Prrx1 knockdown on stemness, migration, and drug resistance of the cell line was assessed, including involvement of SDF-1/CXCR4 signalling. Promising clinical applications of an inhibitor of STAT3 tyrosine phosphorylation, C188–9, and specific blockade with CXCR4 antibody were explored. Results The number of mesenchymal CTCs in blood was closely associated with tumour recurrence or metastasis. Pre-metastatic niche-derived SDF-1 could downregulate Prrx1, which induced the stemness, drug resistance, and increased expression of CXCR4 in HCC cells through the STAT3 pathway in vitro. In vivo, mice bearing tumours of Prrx1 low-expressing cells had significantly shorter survival. In xenograft tumours and clinical samples, loss of Prrx1 was negatively correlated with increased expression of CXCR4 in lung metastatic sites compared with that in the primary foci. Conclusions These findings demonstrate that decreased expression of Prrx1 stimulates SDF-1/CXCR4 signalling and contributes to organ colonisation with blood CTCs in HCC. STAT3 inhibition and specific blockade of CXCR4 have clinical potential as therapeutics for eliminating organ metastasis in advanced HCC.
Collapse
Affiliation(s)
- Yujun Tang
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yishi Lu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuan Chen
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Luo
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Cai
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Bangjian Peng
- Department of Hepatobiliary Surgery, the Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Wenbin Huang
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hangyu Liao
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Liang Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China. .,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China. .,Department of Hepatobiliary Surgery, the Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China.
| | - Mingxin Pan
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
23
|
Yunus M, Jansson PJ, Kovacevic Z, Kalinowski DS, Richardson DR. Tumor-induced neoangiogenesis and receptor tyrosine kinases - Mechanisms and strategies for acquired resistance. Biochim Biophys Acta Gen Subj 2019; 1863:1217-1225. [PMID: 31029846 DOI: 10.1016/j.bbagen.2019.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/23/2019] [Accepted: 04/23/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Angiogenesis is essential for tumor growth, proliferation and metastasis. Tumor-related angiogenesis is complex and involves multiple signaling pathways. Controlling angiogenesis is a promising strategy for limiting cancer progression. SCOPE OF REVIEW Several receptor tyrosine kinases influence the angiogenic response via multiple signaling molecules and pathways. Understanding the functional interaction of kinases in the angiogenic process and development of resistance to kinase inhibition is essential for future successful therapeutic strategies. MAJOR CONCLUSIONS Strategies that target receptor tyrosine kinases and other tumor microenvironment factors simultaneously, or sequentially, are required for achieving an efficient and robust anti-angiogenic response. GENERAL SIGNIFICANCE Understanding the molecular mechanism of angiogenesis has improved, and has led, to the clinical development and approval of anti-angiogenic drugs. While many patients have benefited from these agents, their limited efficacy and the development of resistance remains a challenge. This review highlights current therapies and challenges associated with targeting angiogenesis in cancer.
Collapse
Affiliation(s)
- Madiha Yunus
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), University of Sydney, New South Wales 2006, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), University of Sydney, New South Wales 2006, Australia
| | - Zaklina Kovacevic
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), University of Sydney, New South Wales 2006, Australia
| | - Danuta S Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), University of Sydney, New South Wales 2006, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), University of Sydney, New South Wales 2006, Australia.
| |
Collapse
|
24
|
Jones KM, Karanam B, Jones-Triche J, Sandey M, Henderson HJ, Samant RS, Temesgen S, Yates C, Bedi D. Phage Ligands for Identification of Mesenchymal-Like Breast Cancer Cells and Cancer-Associated Fibroblasts. Front Oncol 2019; 8:625. [PMID: 30619759 PMCID: PMC6304394 DOI: 10.3389/fonc.2018.00625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 12/03/2018] [Indexed: 11/18/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is believed to be crucial for primary tumors to escape their original residence and invade and metastasize. To properly define EMT, there is a need for ligands that can identify this phenomenon in tumor tissue and invivo. A phage-display selection screening was performed to select novel binding phage peptides for identification of EMT in breast cancer. Epithelial breast cancer cell line, MCF-7 was transformed to mesenchymal phenotype by TGF-β treatment and was used for selection. Breast fibroblasts were used for subtractive depletion and breast cancer metastatic cell lines MDA-MB-231, T47D-shNMI were used for specificity assay. The binding peptides were identified, and their binding capacities were confirmed by phage capture assay, phage-based ELISA, immunofluorescence microscopy. The phage peptide bearing the 7-amino acid sequence, LGLRGSL, demonstrated selective binding to EMT phenotypic cells (MCF-7/TGF-β and MDA-MB-231) as compared to epithelial subtype, MCF-7, T47D and breast fibroblasts (Hs578T). The selected phage was also able to identify metastatic breast cancer tumor in breast cancer tissue microarray (TMA). These studies suggest that the selected phage peptide LGLRGSL identified by phage-display library, showed significant ability to bind to mesenchymal-like breast cancer cells/ tissues and can serve as a novel probe/ligand for metastatic breast cancer diagnostic and imaging.
Collapse
Affiliation(s)
- Kelvin M Jones
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, United States
| | - Balasubramanyam Karanam
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, United States
| | | | - Maninder Sandey
- Department of Pathobiology, Auburn University, Auburn, AL, United States
| | - Henry J Henderson
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, United States
| | - Rajeev S Samant
- Department of Pathobiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Samuel Temesgen
- Department of Pathobiology, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, United States
| | - Clayton Yates
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, United States
| | - Deepa Bedi
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, United States
| |
Collapse
|
25
|
Li Z, Riesenberg B, Metelli A, Li A, Wu BX. The Role of Platelets in Tumor Growth, Metastasis, and Immune Evasion. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00030-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Xu J, Xie L, Guo W. PDGF/PDGFR effects in osteosarcoma and the "add-on" strategy. Clin Sarcoma Res 2018; 8:15. [PMID: 30083310 PMCID: PMC6071404 DOI: 10.1186/s13569-018-0102-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/18/2018] [Indexed: 01/12/2023] Open
Abstract
New treatment options for advanced osteosarcoma have remained limited. The platelet-derived growth factor (PDGF)/platelet-derived growth factor receptor (PDGFR) pathway plays an important role in the development and metastasis of osteosarcoma, via either direct autocrine stimulation of tumor cells, or paracrine stimulation on tumor stromal cells. It promotes angiogenesis to overcome hypoxia in the tumor microenvironment, and modulates tumor interstitial fluid pressure to control the influx and efflux of other agents. Targeting the PDGF/PDGFR pathway is a promising therapeutic method to overcome drug resistance and improve patients' outcome in osteosarcoma. Further evidence is needed to define the detailed mechanism. Results from clinical trials using PDGF/PDGFR inhibitor as a single agent were disappointing, both in osteosarcoma and soft tissue sarcoma. However, when combined with other agents, named as "add-on" strategy, a synergistic antitumor effect has been confirmed in soft tissue sarcoma, and should be attempted in osteosarcoma.
Collapse
Affiliation(s)
- Jie Xu
- Peking University People's Hospital, Beijing, 100044 China
| | - Lu Xie
- Peking University People's Hospital, Beijing, 100044 China
| | - Wei Guo
- Peking University People's Hospital, Beijing, 100044 China
| |
Collapse
|
27
|
Liu Y, Starr MD, Brady JC, Rushing C, Pang H, Adams B, Alvarez D, Theuer CP, Hurwitz HI, Nixon AB. Modulation of Circulating Protein Biomarkers in Cancer Patients Receiving Bevacizumab and the Anti-Endoglin Antibody, TRC105. Mol Cancer Ther 2018; 17:2248-2256. [DOI: 10.1158/1535-7163.mct-17-0916] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/23/2018] [Accepted: 07/06/2018] [Indexed: 11/16/2022]
|
28
|
Paragangliomas arise through an autonomous vasculo-angio-neurogenic program inhibited by imatinib. Acta Neuropathol 2018; 135:779-798. [PMID: 29305721 PMCID: PMC5904229 DOI: 10.1007/s00401-017-1799-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/16/2017] [Accepted: 12/21/2017] [Indexed: 12/26/2022]
Abstract
Tumours can be viewed as aberrant tissues or organs sustained by tumorigenic stem-like cells that engage into dysregulated histo/organogenetic processes. Paragangliomas, prototypical organoid tumours constituted by dysmorphic variants of the vascular and neural tissues found in normal paraganglia, provide a model to test this hypothesis. To understand the origin of paragangliomas, we built a biobank comprising 77 cases, 18 primary cultures, 4 derived cell lines, 80 patient-derived xenografts and 11 cell-derived xenografts. We comparatively investigated these unique complementary materials using morphofunctional, ultrastructural and flow cytometric assays accompanied by microRNA studies. We found that paragangliomas contain stem-like cells with hybrid mesenchymal/vasculoneural phenotype, stabilized and expanded in the derived cultures. The viability and growth of such cultures depended on the downregulation of the miR-200 and miR-34 families, which allowed high PDGFRA and ZEB1 protein expression levels. Both tumour tissue- and cell culture-derived xenografts recapitulated the vasculoneural paraganglioma structure and arose from mesenchymal-like cells through a fixed developmental sequence. First, vasculoangiogenesis organized the microenvironment, building a perivascular niche which in turn supported neurogenesis. Neuroepithelial differentiation was associated with severe mitochondrial dysfunction, not present in cultured paraganglioma cells, but acquired in vivo during xenograft formation. Vasculogenesis was the Achilles’ heel of xenograft development. In fact, imatinib, that targets endothelial-mural signalling, blocked paraganglioma xenograft formation (11 xenografts from 12 cell transplants in the control group versus 2 out of 10 in the treated group, P = 0.0015). Overall our key results were unaffected by the SDHx gene carrier status of the patient, characterized for 70 out of 77 cases. In conclusion, we explain the biphasic vasculoneural structure of paragangliomas and identify an early and pharmacologically actionable phase of paraganglioma organization.
Collapse
|
29
|
Ham SL, Thakuri PS, Plaster M, Li J, Luker KE, Luker GD, Tavana H. Three-dimensional tumor model mimics stromal - breast cancer cells signaling. Oncotarget 2017; 9:249-267. [PMID: 29416611 PMCID: PMC5787462 DOI: 10.18632/oncotarget.22922] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/09/2017] [Indexed: 12/11/2022] Open
Abstract
Tumor stroma is a major contributor to the biological aggressiveness of cancer cells. Cancer cells induce activation of normal fibroblasts to carcinoma-associated fibroblasts (CAFs), which promote survival, proliferation, metastasis, and drug resistance of cancer cells. A better understanding of these interactions could lead to new, targeted therapies for cancers with limited treatment options, such as triple negative breast cancer (TNBC). To overcome limitations of standard monolayer cell cultures and xenograft models that lack tumor complexity and/or human stroma, we have developed a high throughput tumor spheroid technology utilizing a polymeric aqueous two-phase system to conveniently model interactions of CAFs and TNBC cells and quantify effects on signaling and drug resistance of cancer cells. We focused on signaling by chemokine CXCL12, a hallmark molecule secreted by CAFs, and receptor CXCR4, a driver of tumor progression and metastasis in TNBC. Using three-dimensional stromal-TNBC cells cultures, we demonstrate that CXCL12 – CXCR4 signaling significantly increases growth of TNBC cells and drug resistance through activation of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) pathways. Despite resistance to standard chemotherapy, upregulation of MAPK and PI3K signaling sensitizes TNBC cells in co-culture spheroids to specific inhibitors of these kinase pathways. Furthermore, disrupting CXCL12 – CXCR4 signaling diminishes drug resistance of TNBC cells in co-culture spheroid models. This work illustrates the capability to identify mechanisms of drug resistance and overcome them using our engineered model of tumor-stromal interactions.
Collapse
Affiliation(s)
- Stephanie Lemmo Ham
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Pradip Shahi Thakuri
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Madison Plaster
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Jun Li
- Department of Mathematical Sciences, Kent State University, Kent, OH 44242, USA
| | - Kathryn E Luker
- Department of Radiology, Microbiology and Immunology, Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gary D Luker
- Department of Radiology, Microbiology and Immunology, Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| |
Collapse
|
30
|
Lin S, Huang G, Xiao Y, Sun W, Jiang Y, Deng Q, Peng M, Wei X, Ye W, Li B, Lin S, Wang S, Wu Q, Liang Q, Li Y, Zhang X, Wu Y, Liu P, Pei D, Yu F, Wen Z, Yao Y, Wu D, Li P. CD215+ Myeloid Cells Respond to Interleukin 15 Stimulation and Promote Tumor Progression. Front Immunol 2017; 8:1713. [PMID: 29255466 PMCID: PMC5722806 DOI: 10.3389/fimmu.2017.01713] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 11/20/2017] [Indexed: 12/16/2022] Open
Abstract
Interleukin 15 (IL-15) regulates the development, survival, and functions of multiple innate and adaptive immune cells and plays a dual role in promoting both tumor cell growth and antitumor immunity. Here, we demonstrated that the in vivo injection of recombinant human IL-15 (200 µg/kg) or murine IL-15 (3 µg/kg) to tumor-bearing NOD-SCID-IL2Rg−/− (NSI) mice resulted in increased tumor progression and CD45+ CD11b+ Gr-1+ CD215+ cell expansion in the tumors and spleen. In B16F10-bearing C57BL/6 mice model, we found that murine IL-15 has antitumoral effect since the activation and expansion of CD8+ T cells with murine IL-15 treatment. But no enhanced or reduced tumor growth was observed in mice when human IL-15 was used. However, both murine and human IL-15 promote CD45+ CD11b+ Gr-1+ CD215+ cells expansion. In xenograft tumor models, CD215+ myeloid cells, but not CD215− cells, responded to human IL-15 stimulation and promoted tumor growth. Furthermore, we found that human IL-15 mediated insulin-like growth factor-1 production in CD215+ myeloid cells and blocking IGF-1 reduced the tumor-promoting effect of IL-15. Finally, we observed that higher IGF-1 expression is an indicator of poor prognosis among lung adenocarcinoma patients. These findings provide evidence that IL-15 may promote tumor cell progression via CD215+ myeloid cells, and IGF-1 may be an important candidate that IL-15 facilitates tumor growth.
Collapse
Affiliation(s)
- Shouheng Lin
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Guohua Huang
- Department of Respiratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiren Xiao
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wei Sun
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuchuan Jiang
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiuhua Deng
- Department of Respiratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Muyun Peng
- Department of Thoracic Oncology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xinru Wei
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wei Ye
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Baiheng Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Simiao Lin
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Suna Wang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qiting Wu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qiubin Liang
- Guangdong Zhaotai InVivo Biomedicine Co. Ltd., Guangzhou, China
| | - Yangqiu Li
- Medical College, Institute of Hematology, Jinan University, Guangzhou, China
| | - Xuchao Zhang
- Guangdong Lung Cancer Institute, Medical Research Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yilong Wu
- Guangdong Lung Cancer Institute, Medical Research Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Pentao Liu
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Duanqing Pei
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Fenglei Yu
- Department of Thoracic Oncology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhesheng Wen
- Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yao Yao
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Donghai Wu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Peng Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
31
|
Östman A. PDGF receptors in tumor stroma: Biological effects and associations with prognosis and response to treatment. Adv Drug Deliv Rev 2017; 121:117-123. [PMID: 28970051 DOI: 10.1016/j.addr.2017.09.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/17/2017] [Accepted: 09/27/2017] [Indexed: 12/31/2022]
Abstract
Platelet-derived growth factor (PDGF) ligands and their receptors (PDGFRα and PDGFRβ) regulate mesenchymal cells, such as fibroblasts and pericytes. These cells are important constituents of tumor stroma where they impact on tumor growth, metastasis and drug response. Studies in model systems have demonstrated ability of the PDGF system to regulate the tumor-stimulatory effects of fibroblasts, as well as their ability to promote cancer cell migration and invasion. Animal studies imply PDGFR-signaling as a regulator of tumor drug uptake. Emerging correlative analyses of different tumor collections are identifying clinically relevant variations in stromal PDGFR status, and associations between PDGFR status in tumor stroma and survival. These associations could either relate to effects of stromal PDGFR signaling on the natural course of the disease or response to treatment. The availability of clinically approved PDGFR-inhibitory drugs suggest interesting possibilities for novel clinical studies, performed on selected patient sub-groups, which further exploits tumor stroma-derived PDGFR signaling.
Collapse
|
32
|
Fan KJ, Yang B, Liu Y, Tian XD, Wang B. Inhibition of human lung cancer proliferation through targeting stromal fibroblasts by dihydromyricetin. Mol Med Rep 2017; 16:9758-9762. [PMID: 29039563 DOI: 10.3892/mmr.2017.7802] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/10/2017] [Indexed: 11/06/2022] Open
Abstract
In the present study, the effects of dihydromyricetin on the proliferative potential of fibroblasts and lung carcinoma cells were investigated. Markedly higher expression levels of smooth muscle actin and platelet derived growth factors (PDGFs) were observed in the fibroblasts using reverse transcription-polymerase chain reaction analysis. The expression levels of PDGF-A and PDGF-B were also higher in the lung cancer cells. Western blot analysis revealed higher expression levels of the receptor for platelet-derived growth factor (PDGFRβ) in the lysates from fibroblasts obtained from normal tissues and carcinoma tissues. Treatment of the fibroblasts with dihydromyricetin inhibited the expression of PDGFRβ when treated with a 10 µM concentration for 48 h. Treatment of the fibroblasts with a 10 µM concentration of dihydromyricetin for 48 h led to complete inhibition of the activation of extracellular signal-regulated kinase (Erk)1/2 and Akt. The results of an MTT assay showed that treatment of the fibroblasts with dihydromyricetin significantly reduced the PDGF-mediated increase in the rate of proliferation. The rate of proliferation of the A549 lung cancer cells cultured with fibroblasts was markedly increased, compared with that of the A549 cells cultured alone. However, dihydromyricetin significantly (P<0.05) inhibited the proliferation rate of the A549 cells cultured with fibroblasts, compared with the untreated cultures. The proliferation rates of the A549 cancer cells, A549 cells cultured with fibroblasts, and A549 cells cultured with fibroblasts and treated with dihydromyricetin were found to be were 78.45, 98.45 and 21.37%, respectively. Dihydromyricetin inhibited the proliferative potential of fibroblasts in the lung cancer cells through targeting the activation of Erk1/2 and Akt. Therefore, there is scope for dihydromyricetin to be evaluated further for the treatment of lung cancer.
Collapse
Affiliation(s)
- Kai-Jie Fan
- Department of Thoracic Surgery, PLA General Hospital, Beijing 100853, P.R. China
| | - Bo Yang
- Department of Thoracic Surgery, PLA General Hospital, Beijing 100853, P.R. China
| | - Yang Liu
- Department of Thoracic Surgery, PLA General Hospital, Beijing 100853, P.R. China
| | - Xiao-Dong Tian
- Department of Thoracic Surgery, PLA General Hospital, Beijing 100853, P.R. China
| | - Bo Wang
- Department of Thoracic Surgery, PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
33
|
Leiss L, Mutlu E, Øyan A, Yan T, Tsinkalovsky O, Sleire L, Petersen K, Rahman MA, Johannessen M, Mitra SS, Jacobsen HK, Talasila KM, Miletic H, Jonassen I, Li X, Brons NH, Kalland KH, Wang J, Enger PØ. Tumour-associated glial host cells display a stem-like phenotype with a distinct gene expression profile and promote growth of GBM xenografts. BMC Cancer 2017; 17:108. [PMID: 28173797 PMCID: PMC5294893 DOI: 10.1186/s12885-017-3109-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 02/03/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Little is known about the role of glial host cells in brain tumours. However, supporting stromal cells have been shown to foster tumour growth in other cancers. METHODS We isolated stromal cells from patient-derived glioblastoma (GBM) xenografts established in GFP-NOD/scid mice. With simultaneous removal of CD11b+ immune and CD31+ endothelial cells by fluorescence activated cell sorting (FACS), we obtained a population of tumour-associated glial cells, TAGs, expressing markers of terminally differentiaed glial cell types or glial progenitors. This cell population was subsequently characterised using gene expression analyses and immunocytochemistry. Furthermore, sphere formation was assessed in vitro and their glioma growth-promoting ability was examined in vivo. Finally, the expression of TAG related markers was validated in human GBMs. RESULTS TAGs were highly enriched for the expression of glial cell proteins including GFAP and myelin basic protein (MBP), and immature markers such as Nestin and O4. A fraction of TAGs displayed sphere formation in stem cell medium. Moreover, TAGs promoted brain tumour growth in vivo when co-implanted with glioma cells, compared to implanting only glioma cells, or glioma cells and unconditioned glial cells from mice without tumours. Genome-wide microarray analysis of TAGs showed an expression profile distinct from glial cells from healthy mice brains. Notably, TAGs upregulated genes associated with immature cell types and self-renewal, including Pou3f2 and Sox2. In addition, TAGs from highly angiogenic tumours showed upregulation of angiogenic factors, including Vegf and Angiopoietin 2. Immunohistochemistry of three GBMs, two patient biopsies and one GBM xenograft, confirmed that the expression of these genes was mainly confined to TAGs in the tumour bed. Furthermore, their expression profiles displayed a significant overlap with gene clusters defining prognostic subclasses of human GBMs. CONCLUSIONS Our data demonstrate that glial host cells in brain tumours are functionally distinct from glial cells of healthy mice brains. Furthermore, TAGs display a gene expression profile with enrichment for genes related to stem cells, immature cell types and developmental processes. Future studies are needed to delineate the biological mechanisms regulating the brain tumour-host interplay.
Collapse
Affiliation(s)
- Lina Leiss
- Neuro Clinic, Haukeland University Hospital, Bergen, Norway.,Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ercan Mutlu
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway.
| | - Anne Øyan
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Microbiology and Immunology, Haukeland University Hospital, Bergen, Norway
| | - Tao Yan
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China.,Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Oleg Tsinkalovsky
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Linda Sleire
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Kjell Petersen
- Computational Biology Unit, Uni Computing, Uni Research AS, Bergen, Norway
| | - Mohummad Aminur Rahman
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Mireille Johannessen
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Sidhartha S Mitra
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Hege K Jacobsen
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Krishna M Talasila
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Hrvoje Miletic
- Translational Cancer Research Group, Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Clinical Medicine, Haukeland University Hospital, Bergen, Norway
| | - Inge Jonassen
- Computational Biology Unit, Uni Computing, Uni Research AS, Bergen, Norway.,Department of Informatics, University of Bergen, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China.,Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Nicolaas H Brons
- Core Facility Flow Cytometry, Centre de Recherche Public de la Santé (CRP-Santé), L-1526, Luxembourg, Luxembourg
| | - Karl-Henning Kalland
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Microbiology and Immunology, Haukeland University Hospital, Bergen, Norway
| | - Jian Wang
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China.,Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Per Øyvind Enger
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Neurosurgery, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
34
|
Chia PL, Russell PA, Scott AM, John T. Targeting the vasculature: anti-angiogenic agents for malignant mesothelioma. Expert Rev Anticancer Ther 2016; 16:1235-1245. [DOI: 10.1080/14737140.2016.1244008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Puey Ling Chia
- Department of Medical Oncology, Austin Health, Melbourne, Australia
- Olivia-Newton John Cancer Research Institute, Austin Health, Melbourne, Australia
| | - Prudence A. Russell
- Department of Anatomical Pathology, St. Vincent’s Hospital, University of Melbourne, Melbourne, Australia
| | - Andrew M Scott
- Olivia-Newton John Cancer Research Institute, Austin Health, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia
- Faculty of Medicine, University of Melbourne, Melbourne, Australia
| | - Thomas John
- Department of Medical Oncology, Austin Health, Melbourne, Australia
- Olivia-Newton John Cancer Research Institute, Austin Health, Melbourne, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Australia
| |
Collapse
|
35
|
Carbognin L, Pilotto S, Peretti U, Tortora G, Bria E. Platelet-derived growth factor receptor inhibitors for non-small cell lung cancer: is the odyssey over? Expert Opin Investig Drugs 2016; 25:635-8. [PMID: 27054716 DOI: 10.1080/13543784.2016.1176140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Luisa Carbognin
- a Medical Oncology , University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Sara Pilotto
- a Medical Oncology , University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Umberto Peretti
- a Medical Oncology , University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Giampaolo Tortora
- a Medical Oncology , University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| | - Emilio Bria
- a Medical Oncology , University of Verona, Azienda Ospedaliera Universitaria Integrata , Verona , Italy
| |
Collapse
|
36
|
GZD856, a novel potent PDGFRα/β inhibitor, suppresses the growth and migration of lung cancer cells in vitro and in vivo. Cancer Lett 2016; 375:172-178. [PMID: 26940138 DOI: 10.1016/j.canlet.2016.02.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/28/2016] [Accepted: 02/09/2016] [Indexed: 12/20/2022]
Abstract
Platelet-derived growth factor receptors (PDGFRα/β) play critical roles in the autocrine-stimulated growth and recruitment of cancer-associated fibroblasts (CAFs) of human lung cancer cells. We have identified GZD856 as a new PDGFR inhibitor that potently inhibits PDGFRα/β kinase activity and blocks this signaling pathway in lung cancer cells both in vitro and in vivo. GZD856 strongly suppresses the proliferation of PDGFRα-amplified H1703 (PDGFRβ(-)) human lung cancer cells and demonstrates significant in vivo antitumor efficacy in a xenograft mouse model. Although GZD856 displays only limited in vitro antiproliferative efficiency against PDGFRα(-)/PDGFRβ(+) A549 lung cancer cells, it efficiently inhibits the in vivo growth and metastasis of A549 cancer cells in xenograft and orthotopic models, respectively. The promising in vivo antitumor activity of GZD856 in A549 models may result from its suppression of PDGFR-related microenvironment factors, such as recruitment of CAFs and collagen content in stromal cells. GZD856 may be considered as a promising new candidate for anti-lung cancer drug development.
Collapse
|
37
|
Lappano R, Rosano C, Pisano A, Santolla MF, De Francesco EM, De Marco P, Dolce V, Ponassi M, Felli L, Cafeo G, Kohnke FH, Abonante S, Maggiolini M. A calixpyrrole derivative acts as an antagonist to GPER, a G-protein coupled receptor: mechanisms and models. Dis Model Mech 2015; 8:1237-46. [PMID: 26183213 PMCID: PMC4610237 DOI: 10.1242/dmm.021071] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 07/07/2015] [Indexed: 12/11/2022] Open
Abstract
Estrogens regulate numerous pathophysiological processes, mainly by binding to and activating estrogen receptor (ER)α and ERβ. Increasing amounts of evidence have recently demonstrated that G-protein coupled receptor 30 (GPR30; also known as GPER) is also involved in diverse biological responses to estrogens both in normal and cancer cells. The classical ER and GPER share several features, including the ability to bind to identical compounds; nevertheless, some ligands exhibit opposed activity through these receptors. It is worth noting that, owing to the availability of selective agonists and antagonists of GPER for research, certain differential roles elicited by GPER compared with ER have been identified. Here, we provide evidence on the molecular mechanisms through which a calixpyrrole derivative acts as a GPER antagonist in different model systems, such as breast tumor cells and cancer-associated fibroblasts (CAFs) obtained from breast cancer patients. Our data might open new perspectives toward the development of a further class of selective GPER ligands in order to better dissect the role exerted by this receptor in different pathophysiological conditions. Moreover, calixpyrrole derivatives could be considered in future anticancer strategies targeting GPER in cancer cells.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende 87036, Italy
| | - Camillo Rosano
- U.O.S. Biopolymers and Proteomics, IST-National Institute for Cancer Research, Genova 16132, Italy
| | - Assunta Pisano
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende 87036, Italy
| | - Maria Francesca Santolla
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende 87036, Italy
| | | | - Paola De Marco
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende 87036, Italy
| | - Vincenza Dolce
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende 87036, Italy
| | - Marco Ponassi
- U.O.S. Biopolymers and Proteomics, IST-National Institute for Cancer Research, Genova 16132, Italy
| | - Lamberto Felli
- U.O.S. Biopolymers and Proteomics, IST-National Institute for Cancer Research, Genova 16132, Italy
| | - Grazia Cafeo
- Department of Chemical Sciences, University of Messina, Messina 98166, Italy
| | | | | | - Marcello Maggiolini
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende 87036, Italy
| |
Collapse
|
38
|
Ogawa N, Inokuchi M, Takagi Y, Sugita H, Kato K, Kojima K, Sugihara K. Clinical significance of platelet derived growth factor-C and -D in gastric cancer. Oncol Lett 2015; 10:3495-3501. [PMID: 26788156 PMCID: PMC4665846 DOI: 10.3892/ol.2015.3758] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 09/04/2015] [Indexed: 02/06/2023] Open
Abstract
Platelet-derived growth factor (PDGF)-C and PDGF-D are frequently upregulated in human cancers and play important roles in tumor progression, angiogenesis and metastasis. However, the distribution, frequency and prognostic value of PDGF-C and PDGF-D expression in gastric cancer have not been clarified. The present study evaluated the association between expression of PDGF-C and PDGF-D, clinicopathological factors and outcomes, in patients with gastric cancer. Gastric adenocarcinoma tumor samples were obtained from 204 patients who underwent curative gastrectomy between 2003 and 2007. The expression of PDGF-C and PDGF-D was analyzed by immunohistochemical staining. High expression of PDGF-C and PDGF-D was detected in 114 (56%) and 151 (74%) tumors, respectively. PDGF-D expression was significantly associated with tumor depth (P=0.039), histopathology (P<0.01), tumor stage (P=0.01) and recurrence (P<0.01), whereas PDGF-C expression correlated only with histopathology (P=0.05). High PDGF-D expression was also associated with significantly shorter relapse-free survival (RFS) time (P<0.01), whilst high PDGF-C expression was associated with marginally, but not significantly, shorter RFS (P=0.10). On multivariate analysis, high PDGF-D expression was determined to be an independent prognostic factor (hazard ratio, 3.3; 95% confidence interval, 1.20–9.4; P=0.02). These findings indicate that high PDGF-D expression is strongly associated with tumor progression, recurrence, distant metastasis and poor outcomes in patients with gastric cancer. PDGF-D may therefore be an independent prognostic factor and a novel therapeutic target.
Collapse
Affiliation(s)
- Norihito Ogawa
- Department of Surgical Oncology, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Mikito Inokuchi
- Department of Surgical Oncology, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Yoko Takagi
- Department of Translational Oncology, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Hirofumi Sugita
- Department of Surgical Oncology, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Keiji Kato
- Department of Surgical Oncology, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Kazuyuki Kojima
- Center for Minimally Invasive Surgery, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Kenichi Sugihara
- Department of Surgical Oncology, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| |
Collapse
|
39
|
Murakami H, Ikeda M, Okusaka T, Inaba Y, Iguchi H, Yagawa K, Yamamoto N. A Phase I study of MEDI-575, a PDGFRα monoclonal antibody, in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol 2015. [PMID: 26223436 DOI: 10.1007/s00280-015-2832-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE MEDI-575 is a fully human monoclonal antibody that selectively binds to platelet-derived growth factor receptor alpha (PDGFRα). This open-label Phase I study assessed the safety and tolerability of MEDI-575 in Japanese patients with advanced solid tumors. METHODS The study comprised two parts: Part A, dose escalation; Part B, dose expansion in patients with hepatocellular cancer. In Part A, patients were enrolled into three cohorts: MEDI-575 was administered intravenously over a 21-day treatment cycle at doses of 9 and 15 mg/kg/week (cohorts 1, 2) and 35 mg/kg/3-weekly (cohort 3). In Part B, MEDI-575 25 mg/kg/3-weekly was administered. Secondary measures included assessment of the maximum tolerated dose, pharmacokinetics, immunogenicity and anti-tumor activity. RESULTS Ten and 12 patients were treated in Parts A and B, respectively. There were no dose-limiting toxicities; the maximum tolerated dose was not determined. Common treatment-related adverse events were fatigue (30%) and decreased appetite (20%) in Part A and decreased appetite (33.3%) in Part B. All treatment-related adverse events were grade 1 or 2 in severity. No patients discontinued MEDI-575 because of an adverse event and there were no patient deaths due to adverse events. MEDI-575 binding with PDGFRα resulted in a dose-dependent increase in PDGF-AA ligand, with plateau levels observed within 2 days and sustained during the dosing interval. None of the patients in Part A or B experienced complete or partial responses to treatment. CONCLUSIONS MEDI-575 once weekly and 3-weekly was well tolerated with a favorable pharmacokinetic profile in Japanese patients with advanced solid tumors. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, NCT01102400.
Collapse
Affiliation(s)
- Haruyasu Murakami
- Division of Thoracic Oncology, Shizuoka Cancer Center, 1007 Shimonagakubo, Nagaizumicho Sunto-Gun, Shizuoka, 411-8777, Japan,
| | | | | | | | | | | | | |
Collapse
|
40
|
Raffaghello L, Dazzi F. Classification and biology of tumour associated stromal cells. Immunol Lett 2015; 168:175-82. [PMID: 26145459 DOI: 10.1016/j.imlet.2015.06.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 06/29/2015] [Indexed: 12/26/2022]
Abstract
Stroma is a fundamental component of the tumour microenvironment whereby it supports malignant cell growth and spreading. It consists of different entities including cells of the immune system, vascular structures and fibroblasts. Much attention has recently been paid to fibroblasts since there is compelling evidence that they orchestrate the recruitment of and educate other cells to promote cancer growth. This review proposes to discuss in detail the nomenclature, origin, and biological functions of the different stromal cells residing in tumours.
Collapse
Affiliation(s)
- Lizzia Raffaghello
- Laboratory of Oncology, Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genoa, Italy.
| | - Francesco Dazzi
- Regenerative & Heamatological Medicine, King's College London, UK
| |
Collapse
|
41
|
Zhao Y, Adjei AA. Targeting Angiogenesis in Cancer Therapy: Moving Beyond Vascular Endothelial Growth Factor. Oncologist 2015; 20:660-73. [PMID: 26001391 DOI: 10.1634/theoncologist.2014-0465] [Citation(s) in RCA: 408] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/06/2015] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Angiogenesis, or the formation of new capillary blood vessels, occurs primarily during human development and reproduction; however, aberrant regulation of angiogenesis is also a fundamental process found in several pathologic conditions, including cancer. As a process required for invasion and metastasis, tumor angiogenesis constitutes an important point of control of cancer progression. Although not yet completely understood, the complex process of tumor angiogenesis involves highly regulated orchestration of multiple signaling pathways. The proangiogenic signaling molecule vascular endothelial growth factor (VEGF) and its cognate receptor (VEGF receptor 2 [VEGFR-2]) play a central role in angiogenesis and often are highly expressed in human cancers, and initial clinical efforts to develop antiangiogenic treatments focused largely on inhibiting VEGF/VEGFR signaling. Such approaches, however, often lead to transient responses and further disease progression because angiogenesis is regulated by multiple pathways that are able to compensate for each other when single pathways are inhibited. The platelet-derived growth factor (PDGF) and PDGF receptor (PDGFR) and fibroblast growth factor (FGF) and FGF receptor (FGFR) pathways, for example, provide potential escape mechanisms from anti-VEGF/VEGFR therapy that could facilitate resumption of tumor growth. Accordingly, more recent treatments have focused on inhibiting multiple signaling pathways simultaneously. This comprehensive review discusses the limitations of inhibiting VEGF signaling alone as an antiangiogenic strategy, the importance of other angiogenic pathways including PDGF/PDGFR and FGF/FGFR, and the novel current and emerging agents that target multiple angiogenic pathways for the treatment of advanced solid tumors. IMPLICATIONS FOR PRACTICE Significant advances in cancer treatment have been achieved with the development of antiangiogenic agents, the majority of which have focused on inhibition of the vascular endothelial growth factor (VEGF) pathway. VEGF targeting alone, however, has not proven to be as efficacious as originally hoped, and it is increasingly clear that there are many interconnected and compensatory pathways that can overcome VEGF-targeted inhibition of angiogenesis. Maximizing the potential of antiangiogenic therapy is likely to require a broader therapeutic approach using a new generation of multitargeted antiangiogenic agents.
Collapse
Affiliation(s)
- Yujie Zhao
- Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Alex A Adjei
- Roswell Park Cancer Institute, Buffalo, New York, USA
| |
Collapse
|
42
|
Noskovičová N, Petřek M, Eickelberg O, Heinzelmann K. Platelet-Derived Growth Factor Signaling in the Lung. From Lung Development and Disease to Clinical Studies. Am J Respir Cell Mol Biol 2015; 52:263-84. [DOI: 10.1165/rcmb.2014-0294tr] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
43
|
Chen SY, Lin JS, Lin HC, Shan YS, Cheng YJ, Yang BC. Dependence of fibroblast infiltration in tumor stroma on type IV collagen-initiated integrin signal through induction of platelet-derived growth factor. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:929-39. [PMID: 25686533 DOI: 10.1016/j.bbamcr.2015.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/05/2015] [Indexed: 02/04/2023]
Abstract
Cancer-associated fibroblasts play a crucial role in accelerating tumor progression, but there is a knowledge gap regarding the chemotactic signal activated in a tumor microenvironment. In this study, the expression of type IV collagen was knocked down using a lentiviral-mediated short hairpin RNA strategy. Although there was no obvious effect on cell growth in vitro, silencing the Col4-α1 gene decreased the tumorigenicity of B16F10 in C57BL/6 mice, which was accompanied by a reduction in the infiltration of alpha-smooth muscle actin-positive (α-SMA+) fibroblasts. Silencing the Col4-α1 gene or disrupting integrin engagement by blocking the antibody reduced the expression of platelet-derived growth factor A (PDGF-A), a potent chemotactic factor for fibroblasts. Furthermore, ectopic expression of the autoclustering integrin mutant significantly stimulated PDGF-A expression in murine B16F10 and human U118MG and Huh7 cells. PDGF-A-specific sh-RNA and neutralizing anti-PDGF-A antibody effectively inhibited the transwell migration of fibroblasts. Adding recombinant PDGF-A back to shCol cell-conditioned media restored the fibroblast-attraction ability indicating that PDGF-A is a major chemotactic factor for fibroblasts in the current study model. The integrin-associated PDGF-A production correlated with the activation of Src and ERK. High type IV collagen staining intensity colocalized with elevated PDGF-A expression was observed in tumor tissues obtained from hepatoma and glioma patients. The integrin signal pathway was activated by collagen engagement through Src and ERK, leading to enhanced PDGF-A production, which serves as a key regulator of fibroblast recruitment.
Collapse
Affiliation(s)
- Sheng-Yi Chen
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan.
| | - Jo-Shi Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| | - Huan-Ching Lin
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan.
| | - Yan-Shen Shan
- Department of Surgeon, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| | - Yu-Jung Cheng
- Department of Physical Therapy, China Medical University, Taichung 40402, Taiwan.
| | - Bei-Chang Yang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan; Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Center for Gene Regulation and Signal Transduction Research, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
44
|
Arrondeau J, Huillard O, Tlemsani C, Cessot A, Boudou-Rouquette P, Blanchet B, Thomas-Schoemann A, Vidal M, Tigaud JM, Durand JP, Alexandre J, Goldwasser F. Investigational therapies up to Phase II which target PDGF receptors: potential anti-cancer therapeutics. Expert Opin Investig Drugs 2015; 24:673-87. [PMID: 25599887 DOI: 10.1517/13543784.2015.1005736] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The platelet-derived growth factor receptor (PDGFR) pathway has important functions in cell growth and, by overexpression or mutation, could also be a driver for tumor development. Moreover, PDGFR is expressed in a tumoral microenvironment and could promote tumorigenesis. With these biological considerations, the PDGFR pathway could be an interesting target for therapeutics. Currently, there are many molecules under development that target the PDGFR pathway in different types of cancer. AREAS COVERED In this review, the authors report the different molecules under development, as well as those approved albeit briefly, which inhibit the PDGFR pathway. Furthermore, the authors summarize their specificities, their toxicities, and their development. EXPERT OPINION Currently, most PDGFR kinase inhibitors are multikinase inhibitors and therefore do not simply target the PDGFR pathway. The development of more specific PDGFR inhibitors could improve drug efficacy. Moreover, selecting tumors harboring mutations or amplifications of PDGFR could improve outcomes associated with the use of these molecules. The authors believe that new technologies, such as kinome arrays or pharmacologic assays, could be of benefit to understanding resistance mechanisms and develop more selective PDGFR inhibitors.
Collapse
Affiliation(s)
- Jennifer Arrondeau
- Paris Descartes University, Cochin Hospital, AP-HP, Medical Oncology Department, Angiogenesis Inhibitors Multidisciplinary Study Group (CERIA) , Paris , France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wang P, Song L, Ge H, Jin P, Jiang Y, Hu W, Geng N. Crenolanib, a PDGFR inhibitor, suppresses lung cancer cell proliferation and inhibits tumor growth in vivo. Onco Targets Ther 2014; 7:1761-8. [PMID: 25328409 PMCID: PMC4196792 DOI: 10.2147/ott.s68773] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Platelet-derived growth factor (PDGF) and its receptors (PDGFR), including PDGFRα and PDGFRβ, play important roles in tumorigenesis, tumor progression, and the regulation of stromal cell function. Constitutive activation of PDGFR signaling, gene rearrangement, and activating mutations of PDGFR have been identified in various types of human tumors and malignancies. PDGFRα and PDGFRβ belong to the family of type III receptor tyrosine kinases and, upon stimulation, activate downstream signaling cascades. Crenolanib is a specific tyrosine kinase inhibitor that targets and inhibits the kinase activity of PDGFR and the FMS-related tyrosine kinase 3. Its clinical efficacy in several human tumors is currently under investigation in Phase II clinical trials. In this study, we examined the potential role of crenolanib in the treatment of non-small-cell lung cancer (NSCLC). Using A549 cells as a model system, we have shown that crenolanib is capable of suppressing proliferation and inducing apoptosis in a dose-dependent manner. Crenolanib-treated cells have reduced migratory activity in response to inducers of chemotaxis. Furthermore, the in vivo antitumor activity of crenolanib was confirmed in an NSCLC xenograft tumor model. Injection of crenolanib significantly inhibited the growth of tumor mass by inducing apoptosis in tumor cells. Our results provide strong evidence supporting the use of crenolanib as a potential therapeutic agent in treating NSCLC. This work sets a foundation for further development of targeted and personalized therapeutics for lung cancer.
Collapse
Affiliation(s)
- Ping Wang
- Department of Respiratory Medicine, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Liqiang Song
- Department of Pathology, School of Basic Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Hui Ge
- Department of Respiratory Medicine, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Pule Jin
- Department of Respiratory Medicine, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Yifang Jiang
- Department of Respiratory Medicine, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Wenxia Hu
- Department of Respiratory Medicine, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Nan Geng
- Department of Respiratory Medicine, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| |
Collapse
|
46
|
Becerra CR, Conkling P, Vogelzang N, Wu H, Hong S, Narwal R, Liang M, Tavakkoli F, Pandya N. A phase I dose-escalation study of MEDI-575, a PDGFRα monoclonal antibody, in adults with advanced solid tumors. Cancer Chemother Pharmacol 2014; 74:917-25. [PMID: 25149088 PMCID: PMC4209236 DOI: 10.1007/s00280-014-2567-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/04/2014] [Indexed: 11/16/2022]
Abstract
Purpose The purpose of the study was to evaluate safety and determine the maximum tolerated dose (MTD) of MEDI-575, a fully human monoclonal antibody that selectively binds to platelet-derived growth factor receptor-α (PDGFRα), in patients with advanced solid tumors. Methods This phase I multicenter, open-label, single-arm study enrolled adults in a 3 + 3 dose escalation design to receive MEDI-575 (3, 6, 9, 12, or 15 mg/kg) once weekly (QW) until toxicity or disease progression occurred. One 0.5-mg/kg dose was given before the first dose in the 3-mg/kg cohort to determine pharmacokinetics (PK) and pharmacodynamics under unsaturated conditions. After completion of dose escalation in the QW cohorts, patients were enrolled in two additional cohorts and received MEDI-575 25 or 35 mg/kg every 3 weeks (Q3W). Secondary measures included assessments of PK, immunogenicity, and antitumor activity. Results A total of 35 patients received MEDI-575 QW (n = 23) or Q3W (n = 12). Most treatment-related adverse events were grade 1 or 2 in severity across all dose levels, with fatigue (n = 12) and nausea (n = 8) being reported most frequently. With no reports of dose-limiting toxicities (DLTs), the MTD was not reached. MEDI-575 exhibited a nonlinear PK profile and increased plasma platelet-derived growth factor-AA levels in a dose-dependent manner with limited immunogenicity. Stable disease was reported as the best tumor response in 9 of 29 evaluable patients; however, no objective responses were reported. Conclusion Administration of MEDI-575 QW or Q3W resulted in a favorable safety profile, including a lack of DLTs, but without evidence of antitumor activity in patients with refractory solid tumors.
Collapse
Affiliation(s)
- Carlos R Becerra
- Sammons Cancer Center, Texas Oncology P.A., 3410 Worth St., Suite 300, Dallas, TX, 75246, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
The prognostic significance of cancer-associated fibroblasts in esophageal squamous cell carcinoma. PLoS One 2014; 9:e99955. [PMID: 24945657 PMCID: PMC4063790 DOI: 10.1371/journal.pone.0099955] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 05/20/2014] [Indexed: 12/20/2022] Open
Abstract
Background Cancer-associated fibroblasts (CAF) are activated fibroblasts in the cancer stroma and play an important role in cancer progression. Some reports have indicated the correlation between the expression of CAF markers and adverse prognosis in several cancers. However, no reports have studied CAF phenotype and its clinical relevance in esophageal squamous cell carcinoma (ESCC). Methods We investigated CAF phenotype of ESCC based on histology and immunohistochemical expressions of five CAF markers such as fibroblast activation protein (FAP), smooth muscle actin (SMA), fibroblast-specific protein-1 (FSP1), platelet-derived growth factor receptor (PDGFRα), and PDGFRβ in 116 ESCC tissue samples. Besides, we also examined the correlation of the CAF phenotype with clinical relevance as well as other cancer-microenvironment related factors. Results Histologically immature CAF phenotype was correlated with poor prognosis (p<0.001) and associated with increased microvessel density, increased tumor associated macrophages, and epithelial to mesenchymal transition. CAF markers were characteristically expressed in stromal fibroblast close to tumor cells and the expression pattern of 5 CAF markers was highly heterogeneous in every individual cases. Of five CAF markers, SMA, FSP1, and PDGFRα were unfavorable prognostic indicators of ESCC. The number of positive CAF markers was greater in ESCC with immature CAFs than in those with mature ones. Conclusions Our results demonstrate that histologic classification of CAF phenotype is a reliable and significant prognostic predictor in ESCC. CAF markers have the potential to be diagnostic and therapeutic targets in ESCC.
Collapse
|
48
|
Son D, Na YR, Hwang ES, Seok SH. Platelet-derived growth factor-C (PDGF-C) induces anti-apoptotic effects on macrophages through Akt and Bad phosphorylation. J Biol Chem 2014; 289:6225-35. [PMID: 24421315 DOI: 10.1074/jbc.m113.508994] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
PDGF-C, which is abundant in the malignant breast tumor microenvironment, plays an important role in cell growth and survival. Because tumor-associated macrophages (TAMs) contribute to cancer malignancy, macrophage survival mechanisms are an attractive area of research into controlling tumor progression. In this study, we investigated PDGF-C-mediated signaling pathways involved in anti-apoptotic effects in macrophages. We found that the human malignant breast cancer cell line MDA-MB-231 produced high quantities of PDGF-C, whereas benign MCF-7 cells did not. Recombinant PDGF-C induced PDGF receptor α chain phosphorylation, followed by Akt and Bad phosphorylation in THP-1-derived macrophages. MDA-MB-231 culture supernatants also activated macrophage PDGF-Rα. PDGF-C prevented staurosporine-induced macrophage apoptosis by inhibiting the activation of caspase-3, -7, -8, and -9 and cleavage of poly(ADP-ribose) polymerase. Finally, TAMs isolated from the PDGF-C knockdown murine breast cancer cell line 4T1 and PDGF-C knockdown MDA-MB-231-derived tumor mass showed higher rates of apoptosis than the respective WT controls. Collectively, our results suggest that tumor cell-derived PDGF-C enhances TAM survival, promoting tumor malignancy.
Collapse
Affiliation(s)
- Dain Son
- From the Department of Microbiology and Immunology, and Institute of Endemic Disease, College of Medicine, Seoul National University, Seoul 110-799, Korea
| | | | | | | |
Collapse
|
49
|
Heldin CH. Targeting the PDGF signaling pathway in tumor treatment. Cell Commun Signal 2013; 11:97. [PMID: 24359404 PMCID: PMC3878225 DOI: 10.1186/1478-811x-11-97] [Citation(s) in RCA: 349] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 12/11/2013] [Indexed: 01/15/2023] Open
Abstract
Platelet-derived growth factor (PDGF) isoforms and PDGF receptors have important functions in the regulation of growth and survival of certain cell types during embryonal development and e.g. tissue repair in the adult. Overactivity of PDGF receptor signaling, by overexpression or mutational events, may drive tumor cell growth. In addition, pericytes of the vasculature and fibroblasts and myofibroblasts of the stroma of solid tumors express PDGF receptors, and PDGF stimulation of such cells promotes tumorigenesis. Inhibition of PDGF receptor signaling has proven to useful for the treatment of patients with certain rare tumors. Whether treatment with PDGF/PDGF receptor antagonists will be beneficial for more common malignancies is the subject for ongoing studies.
Collapse
Affiliation(s)
- Carl-Henrik Heldin
- Ludwig Institute for Cancer Research, Science for life laboratory, Uppsala University, Box 595SE-751 24 Uppsala, Sweden.
| |
Collapse
|
50
|
Yu Y, Xiao CH, Tan LD, Wang QS, Li XQ, Feng YM. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-β signalling. Br J Cancer 2013; 110:724-32. [PMID: 24335925 PMCID: PMC3915130 DOI: 10.1038/bjc.2013.768] [Citation(s) in RCA: 481] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 10/31/2013] [Accepted: 11/15/2013] [Indexed: 01/06/2023] Open
Abstract
Background: Cancer-associated fibroblasts (CAFs) activated by tumour cells are the predominant type of stromal cells in breast cancer tissue. The reciprocal effect of CAFs on breast cancer cells and the underlying molecular mechanisms are not fully characterised. Methods: Stromal fibroblasts were isolated from invasive breast cancer tissues and the conditioned medium of cultured CAFs (CAF-CM) was collected to culture the breast cancer cell lines MCF-7, T47D and MDA-MB-231. Neutralising antibody and small-molecule inhibitor were used to block the transforming growth factor-β (TGF-β) signalling derived from CAF-CM, which effect on breast cancer cells. Results: The stromal fibroblasts isolated from breast cancer tissues showed CAF characteristics with high expression levels of α-smooth muscle actin and SDF1/CXCL12. The CAF-CM transformed breast cancer cell lines into more aggressive phenotypes, including enhanced cell–extracellular matrix adhesion, migration and invasion, and promoted epithelial–mesenchymal transition (EMT). Cancer-associated fibroblasts secreted more TGF-β1 than TGF-β2 and TGF-β3, and activated the TGF-β/Smad signalling pathway in breast cancer cells. The EMT phenotype of breast cancer cells induced by CAF-CM was reversed by blocking TGF-β1 signalling. Conclusion: Cancer-associated fibroblasts promoted aggressive phenotypes of breast cancer cells through EMT induced by paracrine TGF-β1. This might be a common mechanism for acquiring metastatic potential in breast cancer cells with different biological characteristics.
Collapse
Affiliation(s)
- Y Yu
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan-Hu-Xi Road, Tianjin 300060, China
| | - C-H Xiao
- 1] Department of Breast Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China [2] Key Laboratory of Breast Cancer Prevention and Treatment of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - L-D Tan
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan-Hu-Xi Road, Tianjin 300060, China
| | - Q-S Wang
- 1] Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan-Hu-Xi Road, Tianjin 300060, China [2] Key Laboratory of Breast Cancer Prevention and Treatment of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - X-Q Li
- 1] Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan-Hu-Xi Road, Tianjin 300060, China [2] Key Laboratory of Breast Cancer Prevention and Treatment of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Y-M Feng
- 1] Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Huan-Hu-Xi Road, Tianjin 300060, China [2] Key Laboratory of Breast Cancer Prevention and Treatment of the Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| |
Collapse
|