1
|
Mitra Ghosh T, Mazumder S, Davis J, Yadav J, Akinpelu A, Alnaim A, Kumar H, Waliagha R, Church Bird AE, Rais-Bahrami S, Bird RC, Mistriotis P, Mishra A, Yates CC, Mitra AK, Arnold RD. Metronomic Administration of Topotecan Alone and in Combination with Docetaxel Inhibits Epithelial-mesenchymal Transition in Aggressive Variant Prostate Cancers. CANCER RESEARCH COMMUNICATIONS 2023; 3:1286-1311. [PMID: 37476073 PMCID: PMC10355222 DOI: 10.1158/2767-9764.crc-22-0427] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/29/2023] [Accepted: 06/21/2023] [Indexed: 07/22/2023]
Abstract
Prostate cancer is the second leading cause of noncutaneous cancer-related deaths in American men. Androgen deprivation therapy (ADT), radical prostatectomy, and radiotherapy remain the primary treatment for patients with early-stage prostate cancer (castration-sensitive prostate cancer). Following ADT, many patients ultimately develop metastatic castration-resistant prostate cancer (mCRPC). Standard chemotherapy options for CRPC are docetaxel (DTX) and cabazitaxel, which increase median survival, although the development of resistance is common. Cancer stem-like cells possess mesenchymal phenotypes [epithelial-to-mesenchymal transition (EMT)] and play crucial roles in tumor initiation and progression of mCRPC. We have shown that low-dose continuous administration of topotecan (METRO-TOPO) inhibits prostate cancer growth by interfering with key cancer pathway genes. This study utilized bulk and single-cell or whole-transcriptome analysis [(RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq)], and we observed greater expression of several EMT markers, including Vimentin, hyaluronan synthase-3, S100 calcium binding protein A6, TGFB1, CD44, CD55, and CD109 in European American and African American aggressive variant prostate cancer (AVPC) subtypes-mCRPC, neuroendocrine variant (NEPC), and taxane-resistant. The taxane-resistant gene FSCN1 was also expressed highly in single-cell subclonal populations in mCRPC. Furthermore, metronomic-topotecan single agent and combinations with DTX downregulated these EMT markers as well as CD44+ and CD44+/CD133+ "stem-like" cell populations. A microfluidic chip-based cell invasion assay revealed that METRO-TOPO treatment as a single agent or in combination with DTX was potentially effective against invasive prostate cancer spread. Our RNA-seq and scRNA-seq analysis were supported by in silico and in vitro studies, suggesting METRO-TOPO combined with DTX may inhibit oncogenic progression by reducing cancer stemness in AVPC through the inhibition of EMT markers and multiple oncogenic factors/pathways. Significance The utilization of metronomic-like dosing regimens of topotecan alone and in combination with DTX resulted in the suppression of makers associated with EMT and stem-like cell populations in AVPC models. The identification of molecular signatures and their potential to serve as novel biomarkers for monitoring treatment efficacy and disease progression response to treatment efficacy and disease progression were achieved using bulk RNA-seq and single-cell-omics methodologies.
Collapse
Affiliation(s)
- Taraswi Mitra Ghosh
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Division of Urology, Department of Surgery, Mass General Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Suman Mazumder
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Center for Pharmacogenomics and Single-Cell Omics (AUPharmGx), Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Joshua Davis
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Jyoti Yadav
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Ayuba Akinpelu
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, Alabama
| | - Ahmed Alnaim
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Harish Kumar
- Department of Biology and Canter for Cancer Research, Tuskegee University, Tuskegee, Alabama
| | - Razan Waliagha
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Allison E. Church Bird
- Flow Cytometry and High-Speed Cell Sorting Laboratory, Auburn University, Auburn, Alabama
| | - Soroush Rais-Bahrami
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Urology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Radiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| | - R. Curtis Bird
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Panagiotis Mistriotis
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, Alabama
| | - Amarjit Mishra
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Clayton C. Yates
- Department of Biology and Canter for Cancer Research, Tuskegee University, Tuskegee, Alabama
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Amit K. Mitra
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Center for Pharmacogenomics and Single-Cell Omics (AUPharmGx), Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| | - Robert D. Arnold
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| |
Collapse
|
2
|
Aust G, Zheng L, Quaas M. To Detach, Migrate, Adhere, and Metastasize: CD97/ADGRE5 in Cancer. Cells 2022; 11:cells11091538. [PMID: 35563846 PMCID: PMC9101421 DOI: 10.3390/cells11091538] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022] Open
Abstract
Tumorigenesis is a multistep process, during which cells acquire a series of mutations that lead to unrestrained cell growth and proliferation, inhibition of cell differentiation, and evasion of cell death. Growing tumors stimulate angiogenesis, providing them with nutrients and oxygen. Ultimately, tumor cells invade the surrounding tissue and metastasize; a process responsible for about 90% of cancer-related deaths. Adhesion G protein-coupled receptors (aGPCRs) modulate the cellular processes closely related to tumor cell biology, such as adhesion and detachment, migration, polarity, and guidance. Soon after first being described, individual human aGPCRs were found to be involved in tumorigenesis. Twenty-five years ago, CD97/ADGRE5 was discovered to be induced in one of the most severe tumors, dedifferentiated anaplastic thyroid carcinoma. After decades of research, the time has come to review our knowledge of the presence and function of CD97 in cancer. In summary, CD97 is obviously induced or altered in many tumor entities; this has been shown consistently in nearly one hundred published studies. However, its high expression at circulating and tumor-infiltrating immune cells renders the systemic targeting of CD97 in tumors difficult.
Collapse
Affiliation(s)
- Gabriela Aust
- Research Laboratories of the Clinic of Visceral, Transplantation, Thoracic, and Vascular Surgery, Medical School, University Hospital Leipzig, Leipzig University, 04103 Leipzig, Germany;
- Research Laboratories of the Clinic of Orthopedics, Traumatology and Plastic Surgery, Medical School, University Hospital Leipzig, Leipzig University, 04103 Leipzig, Germany;
| | - Leyu Zheng
- Research Laboratories of the Clinic of Orthopedics, Traumatology and Plastic Surgery, Medical School, University Hospital Leipzig, Leipzig University, 04103 Leipzig, Germany;
| | - Marianne Quaas
- Research Laboratories of the Clinic of Visceral, Transplantation, Thoracic, and Vascular Surgery, Medical School, University Hospital Leipzig, Leipzig University, 04103 Leipzig, Germany;
- Correspondence:
| |
Collapse
|
3
|
Chen SY, Lih TSM, Li QK, Zhang H. Comparing Urinary Glycoproteins among Three Urogenital Cancers and Identifying Prostate Cancer-Specific Glycoproteins. ACS OMEGA 2022; 7:9172-9180. [PMID: 35350332 PMCID: PMC8945184 DOI: 10.1021/acsomega.1c05223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Prostate cancer, bladder cancer, and renal cancers are major urogenital cancers. Of which, prostate cancer is the most commonly diagnosed and second leading cause of cancer death for men in the United States. For urogenital cancers, urine is considered as proximate body fluid to the tumor site for developing non-invasiveness tests. However, the specific molecular signatures from different urogenital cancers are needed to relate changes in urine to various cancer detections. Herein, we utilized a previously published C4-Tip and C18/MAX-Tip workflow for enrichment of glycopeptides from urine samples and evaluated urinary glycopeptides for its cancer specificity. We analyzed 66 urine samples from bladder cancer (n = 27), prostate cancer (n = 4), clear cell renal cell carcinoma (ccRCC, n = 3), and benign plastic hyperplasia (BPH, n = 32) and then compared them with a previous publication that reported glycopeptides associated with aggressive prostate cancer (Gleason score ≥ 8). We further demonstrated the cancer specificity of the glycopeptides associated with aggressive prostate cancer. In this study, a total of 33 glycopeptides were identified to be specifically differentially expressed in prostate cancer compared to other urogenital cancer types as well as BPH urines. By cross-comparison with our previous urinary glycoproteomic dataset for aggressive prostate cancer, we reported a total of four glycopeptides from glycoproteins DSC2, MGAM, PIK3IP1, and CD55, commonly identified to be prostate cancer-specific. Together, these results deepen our understanding of the urinary glycoproteins associated with urogenital cancer types and expand our knowledge of the cancer specificity of urinary glycoproteins among urogenital cancer progression.
Collapse
Affiliation(s)
- Shao-Yung Chen
- Department
of Pathology, Johns Hopkins University School
of Medicine, Baltimore 21287-0010, Maryland, United States
- Department
of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore 21218-2625, Maryland, United States
| | - Tung-Shing Mamie Lih
- Department
of Pathology, Johns Hopkins University School
of Medicine, Baltimore 21287-0010, Maryland, United States
| | - Qing Kay Li
- Department
of Pathology, Johns Hopkins University School
of Medicine, Baltimore 21287-0010, Maryland, United States
| | - Hui Zhang
- Department
of Pathology, Johns Hopkins University School
of Medicine, Baltimore 21287-0010, Maryland, United States
- Department
of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore 21218-2625, Maryland, United States
- Department
of Urology, Johns Hopkins University, Baltimore 21287, Maryland, United States
- Department
of Oncology, Johns Hopkins University Baltimore 21205, Maryland, United States
| |
Collapse
|
4
|
Keshavarz M, Sabbaghi A, Miri SM, Rezaeyan A, Arjeini Y, Ghaemi A. Virotheranostics, a double-barreled viral gun pointed toward cancer; ready to shoot? Cancer Cell Int 2020; 20:131. [PMID: 32336951 PMCID: PMC7178751 DOI: 10.1186/s12935-020-01219-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/17/2020] [Indexed: 12/23/2022] Open
Abstract
Compared with conventional cancer treatments, the main advantage of oncolytic virotherapy is its tumor-selective replication followed by the destruction of malignant cells without damaging healthy cells. Accordingly, this kind of biological therapy can potentially be used as a promising approach in the field of cancer management. Given the failure of traditional monitoring strategies (such as immunohistochemical analysis (in providing sufficient safety and efficacy necessary for virotherapy and continual pharmacologic monitoring to track pharmacokinetics in real-time, the development of alternative strategies for ongoing monitoring of oncolytic treatment in a live animal model seems inevitable. Three-dimensional molecular imaging methods have recently been considered as an attractive approach to overcome the limitations of oncolytic therapy. These noninvasive visualization systems provide real-time follow-up of viral progression within the cancer tissue by the ability of engineered oncolytic viruses (OVs) to encode reporter transgenes based on recombinant technology. Human sodium/iodide symporter (hNIS) is considered as one of the most prevalent nuclear imaging reporter transgenes that provides precise information regarding the kinetics of gene expression, viral biodistribution, toxicity, and therapeutic outcomes using the accumulation of radiotracers at the site of transgene expression. Here, we provide an overview of pre-clinical and clinical applications of hNIS-based molecular imaging to evaluate virotherapy efficacy. Moreover, we describe different types of reporter genes and their potency in the clinical trials.
Collapse
Affiliation(s)
- Mohsen Keshavarz
- 1The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ailar Sabbaghi
- 2Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| | | | - Abolhasan Rezaeyan
- 4Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Yaser Arjeini
- 5Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ghaemi
- 6Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
5
|
Yin X, Shen C, Yin Y, Cai Z, Wang J, Zhao Z, Chen X, Chen Z, Chen H, Zhang B. Overexpression of CD55 correlates with tumor progression and poor prognosis in gastric stromal tumors. Onco Targets Ther 2019; 12:4703-4712. [PMID: 31417272 PMCID: PMC6594005 DOI: 10.2147/ott.s195182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 04/13/2019] [Indexed: 02/05/2023] Open
Abstract
Backgrounds: Accumulating evidences have demonstrated that CD55 can protect cells from complement-mediated attack, and is involved in tumor dedifferentiation, migration, invasiveness, and metastasis. However, the role of CD55 in gastrointestinal stromal tumors (GISTs) has not been investigated. Aims: Our study aimed to analyze the expression of CD55 in gastric GISTs and its correlations with clinicopathologic characteristics and prognosis. Materials and methods: A total of 118 gastric GIST patients were included in our study. CD55 expression in GIST tissue samples was evaluated using immunohistochemistry. Cumulative survival was conducted using the Kaplan-Meier method. Cox regression analyses were performed to identify factors associated with progression-free survival (PFS) for patients with gastric GISTs. Results: Of 118 gastric GISTs patients included in our study, 44 (37.3%) were positive for CD55 expression. Positive CD55 expression in gastric GISTs was closely associated with tumor size (13.52±7.35 vs 5.07±1.90 cm, respectively; P<0.001), Ki 67 labeling index (P=0.001), mitotic counts (P=0.005), NIH risk classification (P<0.001), PLR (P<0.001), and metastasis at initial diagnosis (P=0.002). Kaplan-Meier analyses revealed that tumor size (P<0.001), mitotic counts (P<0.001), Ki 67 labeling index (P<0.001), PLR (P<0.001), metastasis at initial diagnosis (P=0.031), and CD55 expression (P<0.001) were statistically significant risk factors affecting PFS of patients with gastric GISTs. Cox multivariate survival analysis showed that mitotic counts, Ki 67 labeling index, and CD55 expression were independent predictors of PFS for gastric GISTs. Conclusion: CD55 may be a potential prognostic marker in gastric GISTs patients.
Collapse
Affiliation(s)
- Xiaonan Yin
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Chaoyong Shen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Zhaolun Cai
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Jian Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Zhou Zhao
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Xin Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Zhixin Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Huijiao Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| |
Collapse
|
6
|
Su Y, Liu Y, Behrens CR, Bidlingmaier S, Lee NK, Aggarwal R, Sherbenou DW, Burlingame AL, Hann BC, Simko JP, Premasekharan G, Paris PL, Shuman MA, Seo Y, Small EJ, Liu B. Targeting CD46 for both adenocarcinoma and neuroendocrine prostate cancer. JCI Insight 2018; 3:121497. [PMID: 30185663 DOI: 10.1172/jci.insight.121497] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/24/2018] [Indexed: 12/25/2022] Open
Abstract
Although initially responsive to androgen signaling inhibitors (ASIs), metastatic castration-resistant prostate cancer (mCRPC) inevitably develops and is incurable. In addition to adenocarcinoma (adeno), neuroendocrine prostate cancer (NEPC) emerges to confer ASI resistance. We have previously combined laser capture microdissection and phage antibody display library selection on human cancer specimens and identified novel internalizing antibodies binding to tumor cells residing in their tissue microenvironment. We identified the target antigen for one of these antibodies as CD46, a multifunctional protein that is best known for negatively regulating the innate immune system. CD46 is overexpressed in primary tumor tissue and CRPC (localized and metastatic; adeno and NEPC), but expressed at low levels on normal tissues except for placental trophoblasts and prostate epithelium. Abiraterone- and enzalutamide-treated mCRPC cells upregulate cell surface CD46 expression. Genomic analysis showed that the CD46 gene is gained in 45% abiraterone-resistant mCRPC patients. We conjugated a tubulin inhibitor to our macropinocytosing anti-CD46 antibody and showed that the resulting antibody-drug conjugate (ADC) potently and selectively kills both adeno and NEPC cell lines in vitro (sub-nM EC50) but not normal cells. CD46 ADC regressed and eliminated an mCRPC cell line xenograft in vivo in both subcutaneous and intrafemoral models. Exploratory toxicology studies of the CD46 ADC in non-human primates demonstrated an acceptable safety profile. Thus, CD46 is an excellent target for antibody-based therapy development, which has potential to be applicable to both adenocarcinoma and neuroendocrine types of mCRPC that are resistant to current treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Rahul Aggarwal
- Department of Medicine.,Helen Diller Family Comprehensive Cancer Center
| | | | | | | | - Jeffry P Simko
- Helen Diller Family Comprehensive Cancer Center.,Department of Pathology
| | | | - Pamela L Paris
- Helen Diller Family Comprehensive Cancer Center.,Department of Urology, and
| | | | - Youngho Seo
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
| | - Eric J Small
- Department of Medicine.,Helen Diller Family Comprehensive Cancer Center.,Department of Urology, and
| | - Bin Liu
- Department of Anesthesia.,Helen Diller Family Comprehensive Cancer Center
| |
Collapse
|
7
|
Wang Y, Liao J, Yang YJ, Wang Z, Qin F, Zhu SM, Zheng H, Wang YP. Effect of membrane-bound complement regulatory proteins on tumor cell sensitivity to complement-dependent cytolysis triggered by heterologous expression of the α-gal xenoantigen. Oncol Lett 2018; 15:9061-9068. [PMID: 29805637 PMCID: PMC5958734 DOI: 10.3892/ol.2018.8478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 02/28/2018] [Indexed: 02/05/2023] Open
Abstract
Engineering malignant cells to express a heterologous α-gal antigen can induce heterograft hyperacute rejection, resulting in complement-dependent cytolysis (CDC) of tumor cells, which has been considered as a novel strategy for antitumor therapy. A549 cells engineered to express Galα1-3Galβ1-4GlcNAc-R (α-gal) epitope exhibited strong resistance to CDC treated by normal human serum (NHS) in a previous study. We hypothesized that the expression of membrane-bound complement regulatory proteins (mCRPs) decay accelerating factor (CD55) and protectin (CD59) influenced the efficacy of the α-gal/NHS-mediated antitumor effect to tumor cells in vitro. The present study confirmed that A549 cells expressed high levels of CD55 and CD59, whereas Lovo cells expressed relatively low levels of these proteins. A549 and Lovo cells transfected with plasmids containing or lacking the α-gal epitope were evaluated for their susceptibility to CDC by NHS and detected using a trypan blue exclusion assay. α-gal-expressing Lovo (Lovo-GT) cells were almost completely killed by α-gal-mediated CDC following incubation with 50% NHS, whereas no cytolysis was observed in α-gal expressing A549 (A549-GT) cells. Abrogating CD55 and CD59 function from A549-GT cells by various concentrations of phosphatidylinositol-specific phospholipase C (PI-PLC) or blocking antibodies increased the susceptibility of cells to CDC, and the survival rate decreased significantly comparing to the controls (P<0.05). The findings of the present study indicated that using the α-gal/NHS system to eliminate tumor cells via inducing the complement cascade reaction might represent a feasible approach for the treatment of cancer. However, high levels of mCRP expression may limit the efficacy of this approach. Therefore, an improved efficacy of cancer cell killing may be achieved by combining strategies of heterologous α-gal expression and mCRP downregulation.
Collapse
Affiliation(s)
- Yu Wang
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Juan Liao
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ya-Jun Yang
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhu Wang
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Feng Qin
- Basic Medical Faculty, Dali Medical College, Dali, Yunnan 671003, P.R. China
| | - Sheng-Ming Zhu
- Department of Oncology, Affiliated Taihe Hospital, Yunyang Medical College, Shiyan, Hubei 442000, P.R. China
| | - Hong Zheng
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan-Ping Wang
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
8
|
Abstract
CD59 has been identified as a glycosylphosphatidylinositol-anchored membrane protein that acts as an inhibitor of the formation of the membrane attack complex to regulate complement activation. Recent studies have shown that CD59 is highly expressed in several cancer cell lines and tumor tissues. CD59 also regulates the function, infiltration and phenotypes of a variety of immune cells in the tumor microenvironment. Herein, we summarized recent advances related to the functions and mechanisms of CD59 in the tumor microenvironment. Therapeutic strategies that seek to modulate the functions of CD59 in the tumor microenvironment could be a promising direction for tumor immunotherapy.
Collapse
Affiliation(s)
- Ronghua Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| |
Collapse
|
9
|
Wang Y, Yang YJ, Wang Z, Liao J, Liu M, Zhong XR, Zheng H, Wang YP. CD55 and CD59 expression protects HER2-overexpressing breast cancer cells from trastuzumab-induced complement-dependent cytotoxicity. Oncol Lett 2017; 14:2961-2969. [PMID: 28928834 PMCID: PMC5588148 DOI: 10.3892/ol.2017.6555] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 04/28/2017] [Indexed: 02/05/2023] Open
Abstract
A large proportion (40-60%) of patients with human epidermal growth factor receptor 2 (HER2)-overexpressing breast cancer do not benefit from trastuzumab treatment, potentially due to the lack of complement-dependent cytotoxicity (CDC) activation. In the present study, the effect of complement decay-accelerating factor (CD55) and CD59 glycoprotein precursor (CD59) expression on trastuzumab-induced CDC in HER2-positive breast cancer cell lines was investigated. The CD55 and CD59-overexpressing and HER2-positive cell lines SK-BR-3 and BT474 were selected for subsequent experiments. Blocking CD55 and CD59 function using targeting monoclonal antibodies significantly enhanced the cell lysis of SK-BR-3 and BT474 cells following treatment with trastuzumab. In addition, following treatment with 0.1 U/ml phosphatidylinositol-specific phospholipase C (PI-PLC) for 1 h, CD55 and CD59 surface expression was significantly decreased, and the cell lysis rate was further enhanced. Treatment of SK-BR-3 cells with short hairpin RNA (shRNA) targeting CD55 and CD59 downregulated CD55 and CD59 expression at the mRNA and protein levels, and resulted in significantly enhanced trastuzumab-induced CDC-dependent lysis. The data from the present study suggested that CD55 and CD59 serve roles in blocking trastuzumab-induced CDC, therefore strategies targeting CD55 and CD59 may overcome breast cancer cell resistance to trastuzumab. The results from the present study may provide a basis for developing suitable, personalized treatment strategies to improve the clinical efficacy of trastuzumab for patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
- Yu Wang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ya-Jun Yang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhu Wang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Juan Liao
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Mei Liu
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Breast Cancer Research Center, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Xiao-Rong Zhong
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hong Zheng
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan-Ping Wang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
10
|
Meng ZW, Liu MC, Hong HJ, Du Q, Chen YL. Expression and prognostic value of soluble CD97 and its ligand CD55 in intrahepatic cholangiocarcinoma. Tumour Biol 2017; 39:1010428317694319. [PMID: 28345461 DOI: 10.1177/1010428317694319] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The incidence rate of intrahepatic cholangiocarcinoma is rising, and treatment options are limited. Therefore, new biological markers of intrahepatic cholangiocarcinoma are needed. Immunohistochemistry and enzyme-linked immunosorbent assay were applied to analyze the expressions of CD97, CD55, and soluble CD97 in 71 patients with intrahepatic cholangiocarcinoma and 10 patients with hepatolithiasis. CD97 and CD55 were not expressed in hepatolithiatic tissues, but positive expression was observed in 76.1% (54/71) and 70.4% (50/71) of intrahepatic cholangiocarcinoma patients. The univariate analyses indicated that the positive expressions of CD97 and CD55 were related to short intrahepatic cholangiocarcinoma survival of patients (both p = 0.001). Furthermore, CD97 and CD55 expressions and biliary soluble CD97 levels were significantly associated with histological grade (p = 0.004, 0.002, and 0.012, respectively), lymph node metastases (p = 0.020, 0.038, and 0.001, respectively), and venous invasion (p = 0.003, 0.002, and 0.001, respectively). The multivariate analyses indicated that lymph node metastases (hazard ratio: 2.407, p = 0.003), positive CD55 expression (hazard ratio: 4.096, p = 0.003), and biliary soluble CD97 levels (hazard ratio: 2.434, p = 0.002) were independent risk factors for the intrahepatic cholangiocarcinoma survival. The receiver operating characteristic (ROC) curve analysis indicated that when the cutoff values of biliary soluble CD97 were 1.15 U/mL, the diagnostic value for predicting lymph node metastasis had a sensitivity of 87.5% and a specificity of 51.3%. For intrahepatic cholangiocarcinoma patient death within 60 months at a cutoff value of 0.940 U/mL, the diagnostic value sensitivity was 89.3% and the specificity was 93.3%. Biliary soluble CD97 may be a new biological marker for early diagnosis, prediction of lymph node metastasis and poor prognosis, and discovery of a therapeutic target.
Collapse
Affiliation(s)
- Ze-Wu Meng
- Department of Hepatobiliary Surgery, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, People’s Republic of China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Min-Chao Liu
- Department of Hepatobiliary Surgery, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, People’s Republic of China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Hai-Jie Hong
- Department of Hepatobiliary Surgery, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, People’s Republic of China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Qiang Du
- Department of Hepatobiliary Surgery, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, People’s Republic of China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Yan-Ling Chen
- Department of Hepatobiliary Surgery, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, People’s Republic of China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People’s Republic of China
| |
Collapse
|
11
|
Immunohistochemical Expression and Prognostic Significance of CD97 and its Ligand DAF in Human Cervical Squamous Cell Carcinoma. Int J Gynecol Pathol 2016; 34:473-9. [PMID: 26107567 DOI: 10.1097/pgp.0000000000000200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Accumulating evidences had demonstrated that the CD97, a member of the epidermal growth factor 7-transmembrane family, and its cellular ligand decay accelerating factor (DAF) both play important roles in tumor dedifferentiation, migration, invasiveness, and metastasis. However, the roles of CD97 and DAF in human cervical squamous cell carcinoma (CSCC) have not been investigated. The purpose of this study was to observe the expression profile of CD97 and DAF in CSCC and evaluate their clinical significance. Immunohistochemistry was used to investigate the expression of CD97 and DAF proteins in 97 patients with CSCC and 53 patients with cervical intraepithelial neoplasia, a precursor lesion of CSCC. CD97 and DAF were absent or only weakly expressed in the normal epithelium of the cervix but were present in 83.5% (81/97) and 90.7% (88/97) of CSCC samples, respectively. Overexpression of CD97 was significantly associated with a high International Federation of Gynecology and Obstetrics stage (P=0.010) and lymph node metastasis (P=0.026). The majority of CSCCs, irrespective of staging/grading classification, displayed strong DAF immunostaining. Kaplan-Meier survival analysis revealed that overexpression of CD97 was associated with a worse prognosis. Multivariate analyses showed that the International Federation of Gynecology and Obstetrics stage (P=0.000), lymph node metastasis (P=0.004), and CD97 expression (P=0.040) were independent risk factors for overall survival. The present study suggested that the expressions of CD97 and DAF were both upregulated in CSCC. The expression level of CD97 in CSCC was associated with the severity of the tumor. Furthermore, CD97 might be an independent poor prognostic factor for CSCC patients.
Collapse
|
12
|
Abstract
Alterations in the homeostasis of several adhesion GPCRs (aGPCRs) have been observed in cancer. The main cellular functions regulated by aGPCRs are cell adhesion, migration, polarity, and guidance, which are all highly relevant to tumor cell biology. Expression of aGPCRs can be induced, increased, decreased, or silenced in the tumor or in stromal cells of the tumor microenvironment, including fibroblasts and endothelial and/or immune cells. For example, ADGRE5 (CD97) and ADGRG1 (GPR56) show increased expression in many cancers, and initial functional studies suggest that both are relevant for tumor cell migration and invasion. aGPCRs can also impact the regulation of angiogenesis by releasing soluble fragments following the cleavage of their extracellular domain (ECD) at the conserved GPCR-proteolytic site (GPS) or other more distal cleavage sites as typical for the ADGRB (BAI) family. Interrogation of in silico cancer databases suggests alterations in other aGPCR members and provides the impetus for further exploration of their potential role in cancer. Integration of knowledge on the expression, regulation, and function of aGPCRs in tumorigenesis is currently spurring the first preclinical studies to examine the potential of aGPCR or the related pathways as therapeutic targets.
Collapse
Affiliation(s)
- Gabriela Aust
- Department of Surgery, Research Laboratories, University of Leipzig, Liebigstraße 19, Leipzig, 04103, Germany.
| | - Dan Zhu
- Department of Neurosurgery and Hematology & Medical Oncology, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Erwin G Van Meir
- Department of Neurosurgery and Hematology & Medical Oncology, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Lei Xu
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| |
Collapse
|
13
|
Murao T, Shiotani A, Fujita Y, Yamanaka Y, Kamada T, Manabe N, Hata J, Nishio K, Haruma K. Overexpression of CD55 from Barrett's esophagus is associated with esophageal adenocarcinoma risk. J Gastroenterol Hepatol 2016; 31:99-106. [PMID: 26202380 DOI: 10.1111/jgh.13055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 06/05/2015] [Accepted: 06/25/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIM Although several molecular biomarkers for esophageal adenocarcinoma (EAC) have been shown to be useful disease indicators, none has been established as a reliable indicator for risk of EAC or have progressed to routine use. The aim was to identify biomarkers of high risk for EAC in patients with Barrett's esophagus (BE). METHODS Following endoscopic observation by magnified endoscopy with narrow band imaging (ME-NBI), brushing was followed by obtaining biopsy samples from columnar-lined esophagus (CLE) and from EAC lesions of EAC patients, and from age- and sex-matched non-EAC controls with BE. Total RNA was extracted for microarray analysis using Affymetrix GeneChip Human Genome U133 plus 2.0 Array. Real-time-PCR analysis of identified candidate genes was used to confirm the results. RESULTS Overall, 9 EAC patients and 50 patients with BE were studied. Seventy-nine candidate genes were identified by microarray analysis based on a proportional hazards model (P < 0.005). Six genes exhibited significantly differential expressions in both BE and cancer lesions of the EAC group compared to BE of the controls. In the brushing samples, median CD55 relative expression levels in cancer lesions were highest and decreased in BE of EAC group and BE of the controls, in that order (P < 0.001). CONCLUSION Over expression of CD55 in brushing samples taken from BE may be associated with the risk of EAC.
Collapse
Affiliation(s)
- Takahisa Murao
- Division of Gastroenterology, Department of Internal Medicine, Japan
| | - Akiko Shiotani
- Division of Gastroenterology, Department of Internal Medicine, Japan
| | - Yoshihiko Fujita
- Department of Genome Biology, Kinki University Faculty of Medicine, Osaka, Japan
| | | | - Tomoari Kamada
- Division of Gastroenterology, Department of Internal Medicine, Japan
| | - Noriaki Manabe
- Department of Clinical Pathology and Laboratory Medicine, Kawasaki Medical School, Kurashiki, Japan
| | - Jiro Hata
- Department of Clinical Pathology and Laboratory Medicine, Kawasaki Medical School, Kurashiki, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kinki University Faculty of Medicine, Osaka, Japan
| | - Ken Haruma
- Division of Gastroenterology, Department of Internal Medicine, Japan
| |
Collapse
|
14
|
He Z, Wu H, Jiao Y, Zheng J. Expression and prognostic value of CD97 and its ligand CD55 in pancreatic cancer. Oncol Lett 2014; 9:793-797. [PMID: 25624904 PMCID: PMC4301556 DOI: 10.3892/ol.2014.2751] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 10/29/2014] [Indexed: 01/27/2023] Open
Abstract
CD97 is a member of the epidermal growth factor-seven transmembrane family. It affects tumor aggressiveness by binding its cellular ligand CD55 and exhibits adhesive properties. Previous studies have shown that CD97 and CD55 are involved in the dedifferentiation, migration, invasiveness and metastasis of tumors. However, little is known regarding the roles of CD97 and CD55 in pancreatic cancer. In this study, immunohistochemistry was used to analyze CD97 and CD55 protein expression in samples obtained from 37 pancreatic cancer patients. CD97 and CD55 were absent or only weakly expressed in the normal pancreatic tissues but strongly expressed in pancreatic cancer tissues (P<0.05), particularly in tissues with lymph node involvement, metastasis or vascular invasion (P<0.05). Notably, CD97 and CD55 were expressed consistently in pancreatic cancer tissues (r2=0.5422; P<0.05). In addition, CD97 and CD55 expression levels were found to significantly correlate with tumor aggressiveness (P<0.01). Multivariate analyses revealed that CD97 and CD55 expression levels were closely associated with prognosis (P<0.05). Taken together, these results indicated that CD97 and its ligand CD55 are upregulated in pancreatic cancers and are closely associated with lymph node involvement, metastasis and vascular invasion. Thus, analysis of both CD97 and CD55 expression may present potential prognostic value for pancreatic cancer.
Collapse
Affiliation(s)
- Zheng He
- Department of General Surgery, The First College of Clinical Medical Sciences, Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Hui Wu
- Department of General Surgery, The First College of Clinical Medical Sciences, Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Yanli Jiao
- Department of General Surgery, The First College of Clinical Medical Sciences, Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Jun Zheng
- Department of General Surgery, The First College of Clinical Medical Sciences, Three Gorges University, Yichang, Hubei 443002, P.R. China
| |
Collapse
|
15
|
LIU MEI, YANG YAJUN, ZHENG HONG, ZHONG XIAORONG, WANG YU, WANG ZHU, WANG YAOGENG, WANG YANPING. Membrane-bound complement regulatory proteins are prognostic factors of operable breast cancer treated with adjuvant trastuzumab: A retrospective study. Oncol Rep 2014; 32:2619-27. [DOI: 10.3892/or.2014.3496] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/20/2014] [Indexed: 11/06/2022] Open
|
16
|
Pio R, Corrales L, Lambris JD. The role of complement in tumor growth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 772:229-62. [PMID: 24272362 DOI: 10.1007/978-1-4614-5915-6_11] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Complement is a central part of the immune system that has developed as a first defense against non-self cells. Neoplastic transformation is accompanied by an increased capacity of the malignant cells to activate complement. In fact, clinical data demonstrate complement activation in cancer patients. On the basis of the use of protective mechanisms by malignant cells, complement activation has traditionally been considered part of the body's immunosurveillance against cancer. Inhibitory mechanisms of complement activation allow cancer cells to escape from complement-mediated elimination and hamper the clinical efficacy of monoclonal antibody-based cancer immunotherapies. To overcome this limitation, many strategies have been developed with the goal of improving complement-mediated effector mechanisms. However, significant work in recent years has identified new and surprising roles for complement activation within the tumor microenvironment. Recent reports suggest that complement elements can promote tumor growth in the context of chronic inflammation. This chapter reviews the data describing the role of complement activation in cancer immunity, which offers insights that may aid the development of more effective therapeutic approaches to control cancer.
Collapse
Affiliation(s)
- Ruben Pio
- Oncology Division (CIMA), and Department of Biochemistry and Genetics (School of Science), University of Navarra, Pamplona, Spain,
| | | | | |
Collapse
|
17
|
Carter D, Lieber A. Protein engineering to target complement evasion in cancer. FEBS Lett 2013; 588:334-40. [PMID: 24239543 DOI: 10.1016/j.febslet.2013.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 11/07/2013] [Accepted: 11/08/2013] [Indexed: 01/31/2023]
Abstract
The complement system is composed of soluble factors in plasma that enhance or "complement" immune-mediated killing through innate and adaptive mechanisms. Activation of complement causes recruitment of immune cells; opsonization of coated cells; and direct killing of affected cells through a membrane attack complex (MAC). Tumor cells up-regulate complement inhibitory factors - one of several strategies to evade the immune system. In many cases as the tumor progresses, dramatic increases in complement inhibitory factors are found on these cells. This review focuses on the classic complement pathway and the role of major complement inhibitory factors in cancer immune evasion as well as on how current protein engineering efforts are being employed to increase complement fixing or to reverse complement resistance leading to better therapeutic outcomes in oncology. Strategies discussed include engineering of antibodies to enhance complement fixation, antibodies that neutralize complement inhibitory proteins as well as engineered constructs that specifically target inhibition of the complement system.
Collapse
Affiliation(s)
- Darrick Carter
- PAI Life Sciences Inc., Seattle, WA, United States; Compliment Corp., Seattle, WA, United States.
| | - André Lieber
- Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
18
|
CD97 amplifies LPA receptor signaling and promotes thyroid cancer progression in a mouse model. Oncogene 2012; 32:2726-38. [PMID: 22797060 DOI: 10.1038/onc.2012.301] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CD97, a member of the adhesion family of G-protein-coupled receptors (GPCRs), complexes with and potentiates lysophosphatidic acid (LPA) receptor signaling to the downstream effector RHOA. We show here that CD97 was expressed in a majority of thyroid cancers but not normal thyroid epithelium and that the level of CD97 expression was further elevated with progression to poorly differentiated and undifferentiated carcinoma. Intratumoral progression also showed that CD97 expression correlates with invasiveness and dedifferentiation. To determine the functional role of CD97, we produced a transgenic model of thyroglobulin promoter-driven CD97 expression. Transgenic CD97 in combination with Thrb(PV), an established mouse model of thyroid follicular cell carcinogenesis, significantly increased the occurrence of vascular invasion and lung metastasis. Expression of transgenic CD97 in thyroid epithelium led to elevated ERK phosphorylation and increased numbers of Ki67+ cells in developing tumors. In addition, tumor cell cultures derived from CD97 transgenic as compared with non-transgenic mice demonstrated enhanced, constitutive and LPA-stimulated ERK activation. In human thyroid cancer cell lines, CD97 depletion reduced RHO-GTP and decreased LPA-stimulated invasion but not EGF-stimulated invasion, further suggesting that CD97 influences an LPA-associated mechanism of progression. Consistent with the above, CD97 expression in human thyroid cancers correlated with LPA receptor and markers of aggressiveness including Ki67 and pAKT. This study shows an autonomous effect of CD97 on thyroid cancer progression and supports the investigation of this GPCR as a therapeutic target for these cancers.
Collapse
|
19
|
Immunohistochemical expression and prognostic value of CD97 and its ligand CD55 in primary gallbladder carcinoma. J Biomed Biotechnol 2012; 2012:587672. [PMID: 22547928 PMCID: PMC3324160 DOI: 10.1155/2012/587672] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 01/25/2012] [Indexed: 12/16/2022] Open
Abstract
Background. CD97 as a member of the EGF-TM7 family with adhesive properties plays an important role in tumor aggressiveness by binding its cellular ligand CD55, which is a complement regulatory protein expressed by cells to protect them from bystander complement attack. Previous studies have shown that CD97 and CD55 both play important roles in tumor dedifferentiation, migration, invasiveness, and metastasis. The aim of this study was to investigate CD97 and CD55 expression in primary gallbladder carcinoma (GBC) and their prognostic significance. Methods. Immunohistochemistry was used to investigate the expression of CD97 and CD55 proteins in 138 patients with GBC. Results. CD97 and CD55 were absent or only weakly expressed in the normal epithelium of the gallbladder but in 69.6% (96/138) and 65.2% (90/138) of GBC, respectively, remarkably at the invasive front of the tumors. In addition, CD97 and CD55 expressions were both significantly associated with high histologic grade (both P = 0.009), advanced pathologic T stage (P = 0.01 and 0.009, resp.) and clinical stage (both P = 0.009), and positive venous/lymphatic invasion (both P = 0.009). Multivariate analyses showed that CD97 (hazard ratio, 3.236; P = 0.02) and CD55 (hazard ratio, 3.209; P = 0.02) expressions and clinical stage (hazard ratio, 3.918; P = 0.01) were independent risk factor for overall survival. Conclusion. Our results provide convincing evidence for the first time that the expressions of CD97 and CD55 are both upregulated in human GBC. The expression levels of CD97 and CD55 in GBC were associated with the severity of the tumor. Furthermore, CD97 and CD55 expressions were independent poor prognostic factors for overall survival in patients with GBC.
Collapse
|
20
|
Ward Y, Lake R, Yin JJ, Heger CD, Raffeld M, Goldsmith PK, Merino M, Kelly K. LPA receptor heterodimerizes with CD97 to amplify LPA-initiated RHO-dependent signaling and invasion in prostate cancer cells. Cancer Res 2011; 71:7301-11. [PMID: 21978933 DOI: 10.1158/0008-5472.can-11-2381] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CD97, an adhesion-linked G-protein-coupled receptor (GPCR), is induced in multiple epithelial cancer lineages. We address here the signaling properties and the functional significance of CD97 expression in prostate cancer. Our findings show that CD97 signals through Gα12/13 to increase RHO-GTP levels. CD97 functioned to mediate invasion in prostate cancer cells, at least in part, by associating with lysophosphatidic acid receptor 1 (LPAR1), leading to enhanced LPA-dependent RHO and extracellular signal-regulated kinase activation. Consistent with its role in invasion, depletion of CD97 in PC3 cells resulted in decreased bone metastasis without affecting subcutaneous tumor growth. Furthermore, CD97 heterodimerized and functionally synergized with LPAR1, a GPCR implicated in cancer progression. We also found that CD97 and LPAR expression were significantly correlated in clinical prostate cancer specimens. Taken together, these findings support the investigation of CD97 as a potential therapeutic cancer target.
Collapse
Affiliation(s)
- Yvona Ward
- Cell and Cancer Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Murakami M, Ugai H, Belousova N, Pereboev A, Dent P, Fisher PB, Everts M, Curiel DT. Chimeric adenoviral vectors incorporating a fiber of human adenovirus 3 efficiently mediate gene transfer into prostate cancer cells. Prostate 2010; 70:362-76. [PMID: 19902467 PMCID: PMC2862273 DOI: 10.1002/pros.21070] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND We have developed a range of adenoviral (Ad) vectors based on human adenovirus serotype 5 (HAdV-5) displaying the fiber shaft and knob domains of species B viruses (HAdV-3, -11, or -35). These species B Ads utilize different cellular receptors than HAdV-5 for infection. We evaluated whether Ad vectors displaying species B fiber shaft and knob domains (Ad5F3Luc1, Ad5F11Luc1, and Ad5F35Luc1) would efficiently infect cancer cells of distinct origins, including prostate cancer. METHODS The fiber chimeric Ad vectors were genetically generated and compared with the original Ad vector (Ad5Luc1) for transductional efficiency in a variety of cancer cell lines, including prostate cancer cells and primary prostate epithelial cells (PrEC), using luciferase as a reporter gene. RESULTS Prostate cancer cell lines infected with Ad5F3Luc1 expressed higher levels of luciferase than Ad5Luc1, as well as the other chimeric Ad vectors. We also analyzed the transductional efficiency via monitoring of luciferase activity in prostate cancer cells when expressed as a fraction of the gene transfer in PrEC cells. In the PC-3 and DU145 cell lines, the gene transfer ratio of cancer cells versus PrEC was once again highest for Ad5F3Luc1. CONCLUSION Of the investigated chimeric HAdV-5/species B vectors, Ad5F3Luc1 was judged to be the most suitable for targeting prostate cancer cells as it showed the highest transductional efficiency in these cells. It is foreseeable that an Ad vector incorporating the HAdV-3 fiber could potentially be used for prostate cancer gene therapy.
Collapse
Affiliation(s)
- Miho Murakami
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Hideyo Ugai
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Natalya Belousova
- Department of Experimental Diagnostic Imaging, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Alexander Pereboev
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Paul Dent
- Department of Biochemistry, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Paul B. Fisher
- Department of Human & Molecular Genetics, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Maaike Everts
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - David T. Curiel
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama
- Correspondence to: David T. Curiel, MD. Ph.D., Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, and the Gene Therapy Center, University of Alabama at Birmingham, 901 19th Street South, BMR2-502, Birmingham, Alabama 35294 USA. Phone: (205) 934-8627. Fax: (205) 975-7476.
| |
Collapse
|
22
|
Noninvasive imaging and radiovirotherapy of prostate cancer using an oncolytic measles virus expressing the sodium iodide symporter. Mol Ther 2009; 17:2041-8. [PMID: 19773744 DOI: 10.1038/mt.2009.218] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Prostate cancer cells overexpress the measles virus (MV) receptor CD46. Herein, we evaluated the antitumor activity of an oncolytic derivative of the MV Edmonston (MV-Edm) vaccine strain engineered to express the human sodium iodide symporter (NIS; MV-NIS virus). MV-NIS showed significant cytopathic effect (CPE) against prostate cancer cell lines in vitro. Infected cells effectively concentrated radioiodide isotopes as measured in vitro by Iodide-125 ((125)I) uptake assays. Virus localization and spread in vivo could be effectively followed by imaging of (123)I uptake. In vivo administration of MV-NIS either locally or systemically (total dose of 9 x 10(6) TCID(50)) resulted in significant tumor regression (P < 0.05) and prolongation of survival (P < 0.01). Administration of (131)I further enhanced the antitumor effect of MV-NIS virotherapy (P < 0.05). In conclusion, MV-NIS is an oncolytic vector with significant antitumor activity against prostate cancer, which can be further enhanced by (131)I administration. The NIS transgene allows viral localization and monitoring by noninvasive imaging which can facilitate dose optimization in a clinical setting.
Collapse
|
23
|
Msaouel P, Galanis E. Measles virotherapy in prostate cancer treatment: a novel antitumor approach. Future Virol 2009. [DOI: 10.2217/fvl.09.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Pavlos Msaouel
- Department of Experimental Physiology, National & Kapodistrian University of Athens, Athens, Greece and, Department of Molecular Medicine, Mayo Clinic, Rochester, 200 First Street SW, Rochester, MN 55905, USA
| | - Evanthia Galanis
- Department of Oncology & Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
24
|
Msaouel P, Iankov ID, Allen C, Morris JC, von Messling V, Cattaneo R, Koutsilieris M, Russell SJ, Galanis E. Engineered measles virus as a novel oncolytic therapy against prostate cancer. Prostate 2009; 69:82-91. [PMID: 18973133 PMCID: PMC2737678 DOI: 10.1002/pros.20857] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND No curative therapy is currently available for locally advanced or metastatic prostate cancer. Oncolytic viruses represent a novel class of therapeutic agents that demonstrates no cross-resistance with existing approaches and can therefore be combined with conventional treatment modalities. Measles virus strains deriving from the Edmonston (MV-Edm) vaccine strain have shown considerable oncolytic activity against a variety of solid tumers and hematologic malignancies. In this study, we investigated the antitumor potential of recombinant MV-Edm derivatives as novel oncolytic agents against prostate cancer. METHODS The susceptibility of prostate cancer cell lines (PC-3, DU-145, and LNCaP) to measles virus infection was demonstrated using an MV-Edm derivative expressing green fluorescent protein (GFP). MV-Edm replication in prostate cancer cell lines was assessed by one step viral growth curves. The oncolytic effect of an MV-Edm strain engineered to express the human carcinoembryonic antigen (CEA) was demonstrated in vitro by MTT assays and in vivo in subcutaneous PC-3 xenografts. CEA levels were quantitated in cell supernatants and mouse serum samples. RESULTS Recombinant MV-Edm strains can effectively infect, replicate in and kill prostate cancer cells. Intratumoral administration of MV-CEA at a total dose of 6 x 10(6) TCID50 resulted in statistically significant tumor growth delay (P = 0.004) and prolongation of survival (P = 0.001) in a subcutaneous PC-3 xenograft model. Viral growth kinetics paralleled CEA production. CONCLUSIONS MV-CEA has potent antitumor activity against prostate cancer cell lines and xenografts. Viral gene expression during treatment can be determined by monitoring of CEA levels in the serum; the latter could allow dose optimization and tailoring of individualized treatment protocols.
Collapse
Affiliation(s)
- Pavlos Msaouel
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Experimental Physiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Ianko D. Iankov
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Cory Allen
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - John C. Morris
- Departmentof Internal Medicine, Division of Endocrinology, Diabetes, Metabolism, Nutrition, Mayo Clinic, Rochester, Minnesota
| | | | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Michael Koutsilieris
- Department of Experimental Physiology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Evanthia Galanis
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
25
|
Varela JC, Atkinson C, Woolson R, Keane TE, Tomlinson S. Upregulated expression of complement inhibitory proteins on bladder cancer cells and anti-MUC1 antibody immune selection. Int J Cancer 2008; 123:1357-63. [PMID: 18561323 PMCID: PMC2681224 DOI: 10.1002/ijc.23676] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Membrane complement inhibitors (CD46, CD55 and CD59) are upregulated in some human cancers indicating that they play a role in immune evasion. We investigated complement inhibitor expression in bladder cancer and examined the hypothesis that selective pressure of an antibody response (anti-MUC1) results in the upregulated expression of complement inhibitors on tumor cells. Paired samples of tumor and normal tissue from 22 bladder cancer patients were analyzed for expression of MUC1, CD46, CD55 and CD59, and matched serum samples analyzed for anti-MUC1 IgM and IgG levels. Relationships between anti-MUC1 antibody levels and complement inhibitor expression were investigated. MUC1 mRNA was upregulated in 86% of tumor samples. CD46 was upregulated in 77%, CD55 in 55% and CD59 in 59% of tumors. Low titer anti-MUC1 IgM was detected in normal human sera, but was elevated in 41% of the bladder cancer patients. Anti-MUC1 IgG was virtually absent from normal sera, but present in 32% of the cancer patients. There was a direct relationship between anti-MUC1 antibody titer and expression level of complement inhibitors. Analysis of the correlation of each antibody with the expression of each complement inhibitor by Spearman's rank test revealed a strong correlation between both anti-MUC1 IgM and IgG levels and increased expression of CD46 and CD55, and combined anti-MUC1 IgM/IgG levels correlated with increased expression of all 3 complement inhibitors. In conclusion, the data demonstrate upregulated complement inhibitor expression and the presence of an anti-MUC1 antibody response in bladder cancer patients and support the hypothesis of antibody-mediated immune selection.
Collapse
Affiliation(s)
- Juan Carlos Varela
- Department of Microbiology and Immunology, Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | |
Collapse
|
26
|
Varela JC, Imai M, Atkinson C, Ohta R, Rapisardo M, Tomlinson S. Modulation of protective T cell immunity by complement inhibitor expression on tumor cells. Cancer Res 2008; 68:6734-42. [PMID: 18701498 PMCID: PMC2681227 DOI: 10.1158/0008-5472.can-08-0502] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Complement-inhibitory proteins expressed on cancer cells can provide protection from antitumor antibodies and may potentially modulate the induction of an immune response to tumor-associated antigens. In the current study, we investigated the consequences of complement inhibitor down-regulation on the effector and inductive phases of an immune response. Stable small interfering RNA-mediated down-regulation of the complement inhibitor Crry on MB49 murine bladder cancer cells increased their susceptibility to monoclonal antibody and complement in vitro. In a syngeneic model of metastatic cancer, the down-regulation of Crry on i.v.-injected MB49 cells was associated with a significant decrease in tumor burden and an increase in the survival of challenged mice. However, monoclonal antibody therapy had no additional benefit. There was an antitumor IgG response, but the response was not effected by Crry down-regulation on inoculated tumor cells. Down-regulation of Crry on MB49 cells resulted in an enhanced antitumor T-cell response in challenged mice (measured by lymphocyte IFN-gamma secretion), and CD8+ T cell depletion of mice prior to injection of MB49 cells completely abrogated the effect of Crry down-regulation on tumor burden and survival. Deficiency of C3 also abrogated the effect of Crry down-regulation on the survival of MB49-challenged mice, indicating a complement-dependent mechanism. These data indicate that complement inhibitors expressed on a tumor cell can suppress a T cell response and that enhancing complement activation on a tumor cell surface can promote protective T cell immunity.
Collapse
Affiliation(s)
- Juan C Varela
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | |
Collapse
|
27
|
Ikeda JI, Morii E, Liu Y, Qiu Y, Nakamichi N, Jokoji R, Miyoshi Y, Noguchi S, Aozasa K. Prognostic Significance of CD55 Expression in Breast Cancer. Clin Cancer Res 2008; 14:4780-6. [DOI: 10.1158/1078-0432.ccr-07-1844] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
28
|
Berry LJ, Au GG, Barry RD, Shafren DR. Potent oncolytic activity of human enteroviruses against human prostate cancer. Prostate 2008; 68:577-87. [PMID: 18288643 DOI: 10.1002/pros.20741] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Oncolytic virotherapy offers a unique treatment modality for prostate cancer, especially stages that are resistant to current therapies, with the additional benefit of preferentially targeting tumor cells amongst an environment of healthy tissue. Herein, the low pathogenic enteroviruses; Coxsackievirus A21 (CVA21), as well as a bio-selected variant of Coxsackievirus A21 (CVA21-DAFv) and Echovirus 1 (EV1) are evaluated as novel oncolytic agents against human prostate cancer. METHODS The surface expression of viral receptors required for enterovirus cell attachment/entry, including intercellular adhesion molecule-1 (ICAM-1), decay-accelerating factor (DAF) and integrin alpha(2)beta(1) on a number of human prostate cancer lines was assessed by flow cytometry. Susceptibility to viral oncolysis was determined via in vitro cell lysis assays performed on cell monolayers cultured in micro titer plates. The in vivo oncolytic efficacy of the enteroviruses was assessed using xenograft models in immune compromised SCID-mice following systemic challenge. RESULTS The majority of prostate cancer lines tested expressed surface ICAM-1 and/or DAF, or alpha(2)beta(1), facilitating significant degrees of oncolysis following in vitro viral challenge. Systemic delivery of each of the three viruses induced reduction of xenograft tumor burdens in vivo, and a therapeutic dose-response was demonstrated for escalating doses of EV1 in the LNCaP animal model. CONCLUSION Enteroviruses CVA21, CVA21-DAFv, and EV1 are potentially potent oncolytic agents against human prostate cancer.
Collapse
Affiliation(s)
- Linda J Berry
- The Picornavirus Research Unit, School of Biomedical Sciences, Faculty of Health, The University of Newcastle, Newcastle, New South Wales, Australia
| | | | | | | |
Collapse
|
29
|
Abstract
Tissue microarrays (TMAs) offer the potential to rapidly translate genomics and basic science research findings to practical clinical application. This is particularly true in the field of cancer biomarker research, where TMAs can be used for candidate biomarker validation and association with patient clinical, pathologic, and outcomes parameters. In this review, we examine the effect of TMA use on prostate cancer biomarker research, focusing on the types of TMAs that have been used, and the biomarkers that have been examined. The results demonstrate that TMAs have been very effective in screening candidate biomarkers for subsequent, extended evaluation in large patient populations. In addition, the use of TMAs in multiple biomarker series allows for the statistical analysis of sets of biomarkers as diagnostic or prognostic tests. The processes used here can be applied to any tumor type to improve patient diagnosis, prognosis, and treatment response prediction.
Collapse
|
30
|
Loberg RD, Day LL, Dunn R, Kalikin LM, Pienta KJ. Inhibition of decay-accelerating factor (CD55) attenuates prostate cancer growth and survival in vivo. Neoplasia 2006; 8:69-78. [PMID: 16533428 PMCID: PMC1584292 DOI: 10.1593/neo.05679] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Decay-accelerating factor (CD55) is a member of membrane-bound complement-regulatory proteins. CD55 expression correlates with poor survival in patients with colorectal cancer and has been implicated in the survival and tumorigenesis of blood-borne malignancies. Histologic analysis of clinical specimens from patients with advanced prostate cancer revealed an increase in CD55 expression in prostate tumor epithelial cells. CD55 was shown to be functionally active and to inhibit complement-mediated lysis in PC-3 and DU145 cells. The percentage of lysis was correlative with the CD55 expression profile observed in these prostate cancer cell lines. These data suggest that CD55 is an important regulator of prostate cancer cell survival. As a result, we have hypothesized that CD55 expression on prostate cancer cells promotes cell survival and contributes to the metastatic potential of prostate cancer cells. To determine the role of CD55 in prostate cancer tumorigenesis and metastasis, we generated PC-3(Luc) prostate cancer cells with CD55 siRNA-targeted disruption. We found that PC-3(Luc)/CD55 siRNA constructs in SCID mice resulted in a significant attenuation of overall tumor burden. Further investigation into the mechanisms of CD55-mediated tumor cell/microenvironment interaction is necessary to understand the role of CD55 in tumor cell survival and metastatic lesion formation.
Collapse
Affiliation(s)
- Robert D Loberg
- Department of Urology, University of Michigan Urology Center, Ann Arbor, MI, USA.
| | | | | | | | | |
Collapse
|