1
|
Nayerpour Dizaj T, Doustmihan A, Sadeghzadeh Oskouei B, Akbari M, Jaymand M, Mazloomi M, Jahanban-Esfahlan R. Significance of PSCA as a novel prognostic marker and therapeutic target for cancer. Cancer Cell Int 2024; 24:135. [PMID: 38627732 PMCID: PMC11020972 DOI: 10.1186/s12935-024-03320-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 03/30/2024] [Indexed: 04/20/2024] Open
Abstract
One of the contributing factors in the diagnosis and treatment of most cancers is the identification of their surface antigens. Cancer tissues or cells have their specific antigens. Some antigens that are present in many cancers elicit different functions. One of these antigens is the prostate stem cell antigen (PSCA) antigen, which was first identified in the prostate. PSCA is a cell surface protein that has different functions in different tissues. It can play an inhibitory role in cell proliferation as well as a tumor-inducing role. PSCA has several genetic variants involved in cancer susceptibility in some tissues, so identifying the characteristics of this antigen and its relationship with clinical features can provide more information on diagnosis and treatment of patients with cancers. Most studies on the PSCA have focused on prostate cancer. While it is also expressed in other cancers, little attention has been paid to its role as a valuable diagnostic, prognostic, and therapeutic tool in other cancers. PSCA has several genetic variants that seem to play a significant role in cancer susceptibility in some tissues, so identifying the characteristics of this antigen and its relationship and variants with clinical features can be beneficial in concomitant cancer therapy and diagnosis, as theranostic tools. In this study, we will review the alteration of the PSCA expression and its polymorphisms and evaluate its clinical and theranostics significance in various cancers.
Collapse
Affiliation(s)
- Tina Nayerpour Dizaj
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Doustmihan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Sadeghzadeh Oskouei
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Akbari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - MirAhmad Mazloomi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Wu W, Huang T, Li S, Gong G, Zhao D, Qiu Y. Subtyping and prognostic model construction based on vesicle-mediated transport-related genes in colorectal cancer. Hum Mol Genet 2024; 33:478-490. [PMID: 37971354 DOI: 10.1093/hmg/ddad197] [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: 09/18/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is impacted by various environmental and genetic variables. Dysregulation of vesicle-mediated transport-related genes (VMTRGs) has been observed in many malignancies, but their effect on prognosis in CRC remains unclear. METHODS CRC samples were clustered into varying subtypes per differential expression of VMTRGs. R package was utilized to explore differences in survival, immune, and drug sensitivity among different disease subtypes. According to differentially expressed genes (DEGs) between subtypes, regression analysis was employed to build a riskscore model and identify independent prognostic factors. The model was validated through a Gene Expression Omnibus (GEO) dataset. Immune landscape, immunophenoscore (IPS), and Tumor Immune Dysfunction and Exclusion (TIDE) scores for different risk groups were calculated. RESULTS Two subtypes of CRC were identified based on VMTRGs, which showed significant differences in survival rates, immune cell infiltration abundance, immune functional activation levels, and immune checkpoint expression levels. Cluster2 exhibited higher sensitivity to anti-tumor drugs such as Nilotinib, Cisplatin, and Oxaliplatin compared to Cluster1. DEGs were mainly enriched in biological processes such as epidermis development, epidermal cell differentiation, and receptor-ligand activity, and signaling pathways like pancreatic secretion. The constructed 13-gene riskscore model demonstrated good predictive ability for CRC patients' prognosis. Furthermore, differences in immune landscape, IPS, and TIDE scores were observed among different risk groups. CONCLUSION This study successfully obtained two CRC subtypes with distinct survival statuses and immune levels based on differential expression of VMTRGs. A 13-gene risk model was constructed. The findings had important implications for prognosis and treatment of CRC.
Collapse
Affiliation(s)
- Wei Wu
- Department of General Surgery, The Central Hospital of Xiaogan, 6 Guangchang Road, Xiaogan City, Hubei Province 432000, China
| | - Tong Huang
- Department of General Surgery, The Central Hospital of Xiaogan, 6 Guangchang Road, Xiaogan City, Hubei Province 432000, China
| | - Shengwen Li
- Department of General Surgery, The Central Hospital of Xiaogan, 6 Guangchang Road, Xiaogan City, Hubei Province 432000, China
| | - Guangwei Gong
- Department of General Surgery, The Central Hospital of Xiaogan, 6 Guangchang Road, Xiaogan City, Hubei Province 432000, China
| | - Dan Zhao
- Department of General Surgery, The Central Hospital of Xiaogan, 6 Guangchang Road, Xiaogan City, Hubei Province 432000, China
| | - Yue Qiu
- Department of General Surgery, The Central Hospital of Xiaogan, 6 Guangchang Road, Xiaogan City, Hubei Province 432000, China
| |
Collapse
|
3
|
Strategies for Isolating and Propagating Circulating Tumor Cells in Men with Metastatic Prostate Cancer. Diagnostics (Basel) 2022; 12:diagnostics12020497. [PMID: 35204587 PMCID: PMC8870963 DOI: 10.3390/diagnostics12020497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/14/2022] [Accepted: 02/11/2022] [Indexed: 11/17/2022] Open
Abstract
Selecting a well-suited method for isolating/characterizing circulating tumor cells (CTCs) is challenging. Evaluating sensitive and specific markers for prostate cancer (PCa)-specific CTC identification and analysis is crucial. We used the CellCollector EpCAM-functionalized system (CC-EpCAM) and evaluated and developed a PCa-functionalized version (CC-PCa); we then compared CTC isolation techniques that exploit the physical and biological properties of CTCs. We established two cohorts of metastatic PCa patients (mPCa; 15 in cohort 1 and 10 in cohort 2). CTC cultivation experiments were conducted with two capturing methods (Ficoll and ScreenCell). The most sensitive detection rates and highest CTC counts were reached with the CC-PCa and ScreenCell system. Patients with ≥5 CTCs isolated with CC-EpCAM had an overall survival (OS) of 0.93 years, and patients with ≥5 CTCs isolated with CC-PCa had an OS of 1.5 years in cohort 1. Nevertheless, we observed the highest sensitivity and specificity for 24-month survival by the Ficoll with CD45 depletion and ScreenCell system with May-Grunwald Giemsa (MGG) staining. The EpCAM molecule is an essential factor related to OS for CTC isolation based on biological properties in mPCa patients. The best-suited CTC capture system is not limited to one characteristic of cells but adapted to downstream analysis.
Collapse
|
4
|
Kumar K, Ghosh A. Radiochemistry, Production Processes, Labeling Methods, and ImmunoPET Imaging Pharmaceuticals of Iodine-124. Molecules 2021; 26:E414. [PMID: 33466827 PMCID: PMC7830191 DOI: 10.3390/molecules26020414] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 01/01/2023] Open
Abstract
Target-specific biomolecules, monoclonal antibodies (mAb), proteins, and protein fragments are known to have high specificity and affinity for receptors associated with tumors and other pathological conditions. However, the large biomolecules have relatively intermediate to long circulation half-lives (>day) and tumor localization times. Combining superior target specificity of mAbs and high sensitivity and resolution of the PET (Positron Emission Tomography) imaging technique has created a paradigm-shifting imaging modality, ImmunoPET. In addition to metallic PET radionuclides, 124I is an attractive radionuclide for radiolabeling of mAbs as potential immunoPET imaging pharmaceuticals due to its physical properties (decay characteristics and half-life), easy and routine production by cyclotrons, and well-established methodologies for radioiodination. The objective of this report is to provide a comprehensive review of the physical properties of iodine and iodine radionuclides, production processes of 124I, various 124I-labeling methodologies for large biomolecules, mAbs, and the development of 124I-labeled immunoPET imaging pharmaceuticals for various cancer targets in preclinical and clinical environments. A summary of several production processes, including 123Te(d,n)124I, 124Te(d,2n)124I, 121Sb(α,n)124I, 123Sb(α,3n)124I, 123Sb(3He,2n)124I, natSb(α, xn)124I, natSb(3He,n)124I reactions, a detailed overview of the 124Te(p,n)124I reaction (including target selection, preparation, processing, and recovery of 124I), and a fully automated process that can be scaled up for GMP (Good Manufacturing Practices) production of large quantities of 124I is provided. Direct, using inorganic and organic oxidizing agents and enzyme catalysis, and indirect, using prosthetic groups, 124I-labeling techniques have been discussed. Significant research has been conducted, in more than the last two decades, in the development of 124I-labeled immunoPET imaging pharmaceuticals for target-specific cancer detection. Details of preclinical and clinical evaluations of the potential 124I-labeled immunoPET imaging pharmaceuticals are described here.
Collapse
Affiliation(s)
- Krishan Kumar
- Laboratory for Translational Research in Imaging Pharmaceuticals, The Wright Center of Innovation in Biomedical Imaging, Department of Radiology, The Ohio State University, Columbus, OH 43212, USA;
| | | |
Collapse
|
5
|
Zhao Z, Li E, Luo L, Zhao S, Liu L, Wang J, Kang R, Luo J. A PSCA/PGRN–NF-κB–Integrin–α4 Axis Promotes Prostate Cancer Cell Adhesion to Bone Marrow Endothelium and Enhances Metastatic Potential. Mol Cancer Res 2019; 18:501-513. [PMID: 31722969 DOI: 10.1158/1541-7786.mcr-19-0278] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/13/2019] [Accepted: 11/07/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Zhigang Zhao
- Department of Urology and Andrology, The First Affiliated Hospital of Guangzhou Medical University; Guangdong Provincial Key Laboratory of Urology, Guangzhou, Guangdong Province, China.
| | - Ermao Li
- Medical school, University of South China, Hengyang, Hunan Province, China
| | - Lianmin Luo
- Department of Urology and Andrology, The First Affiliated Hospital of Guangzhou Medical University; Guangdong Provincial Key Laboratory of Urology, Guangzhou, Guangdong Province, China
| | - Shankun Zhao
- Department of Urology and Andrology, The First Affiliated Hospital of Guangzhou Medical University; Guangdong Provincial Key Laboratory of Urology, Guangzhou, Guangdong Province, China
| | - Luhao Liu
- Department of organ transplantation, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Jiamin Wang
- Department of Urology and Andrology, The First Affiliated Hospital of Guangzhou Medical University; Guangdong Provincial Key Laboratory of Urology, Guangzhou, Guangdong Province, China
| | - Ran Kang
- Department of Urology, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Jintai Luo
- Department of Urology and Andrology, The First Affiliated Hospital of Guangzhou Medical University; Guangdong Provincial Key Laboratory of Urology, Guangzhou, Guangdong Province, China
| |
Collapse
|
6
|
Merrick BA, Phadke DP, Bostrom MA, Shah RR, Wright GM, Wang X, Gordon O, Pelch KE, Auerbach SS, Paules RS, DeVito MJ, Waalkes MP, Tokar EJ. Arsenite malignantly transforms human prostate epithelial cells in vitro by gene amplification of mutated KRAS. PLoS One 2019; 14:e0215504. [PMID: 31009485 PMCID: PMC6476498 DOI: 10.1371/journal.pone.0215504] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/04/2019] [Indexed: 12/20/2022] Open
Abstract
Inorganic arsenic is an environmental human carcinogen of several organs including the urinary tract. RWPE-1 cells are immortalized, non-tumorigenic, human prostate epithelia that become malignantly transformed into the CAsE-PE line after continuous in vitro exposure to 5μM arsenite over a period of months. For insight into in vitro arsenite transformation, we performed RNA-seq for differential gene expression and targeted sequencing of KRAS. We report >7,000 differentially expressed transcripts in CAsE-PE cells compared to RWPE-1 cells at >2-fold change, q<0.05 by RNA-seq. Notably, KRAS expression was highly elevated in CAsE-PE cells, with pathway analysis supporting increased cell proliferation, cell motility, survival and cancer pathways. Targeted DNA sequencing of KRAS revealed a mutant specific allelic imbalance, ‘MASI’, frequently found in primary clinical tumors. We found high expression of a mutated KRAS transcript carrying oncogenic mutations at codons 12 and 59 and many silent mutations, accompanied by lower expression of a wild-type allele. Parallel cultures of RWPE-1 cells retained a wild-type KRAS genotype. Copy number analysis and sequencing showed amplification of the mutant KRAS allele. KRAS is expressed as two splice variants, KRAS4a and KRAS4b, where variant 4b is more prevalent in normal cells compared to greater levels of variant 4a seen in tumor cells. 454 Roche sequencing measured KRAS variants in each cell type. We found KRAS4a as the predominant transcript variant in CAsE-PE cells compared to KRAS4b, the variant expressed primarily in RWPE-1 cells and in normal prostate, early passage, primary epithelial cells. Overall, gene expression data were consistent with KRAS-driven proliferation pathways found in spontaneous tumors and malignantly transformed cell lines. Arsenite is recognized as an important environmental carcinogen, but it is not a direct mutagen. Further investigations into this in vitro transformation model will focus on genomic events that cause arsenite-mediated mutation and overexpression of KRAS in CAsE-PE cells.
Collapse
Affiliation(s)
- B. Alex Merrick
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| | - Dhiral P. Phadke
- Sciome, LLC, Research Triangle Park, North Carolina, United States of America
| | - Meredith A. Bostrom
- David H. Murdock Research Institute, Kannapolis, North Carolina, United States of America
| | - Ruchir R. Shah
- Sciome, LLC, Research Triangle Park, North Carolina, United States of America
| | - Garron M. Wright
- David H. Murdock Research Institute, Kannapolis, North Carolina, United States of America
| | - Xinguo Wang
- David H. Murdock Research Institute, Kannapolis, North Carolina, United States of America
| | - Oksana Gordon
- David H. Murdock Research Institute, Kannapolis, North Carolina, United States of America
| | - Katherine E. Pelch
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Scott S. Auerbach
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Richard S. Paules
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Michael J. DeVito
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Michael P. Waalkes
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Erik J. Tokar
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
7
|
Townsend MH, Shrestha G, Robison RA, O’Neill KL. The expansion of targetable biomarkers for CAR T cell therapy. J Exp Clin Cancer Res 2018; 37:163. [PMID: 30031396 PMCID: PMC6054736 DOI: 10.1186/s13046-018-0817-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/28/2018] [Indexed: 12/13/2022] Open
Abstract
Biomarkers are an integral part of cancer management due to their use in risk assessment, screening, differential diagnosis, prognosis, prediction of response to treatment, and monitoring progress of disease. Recently, with the advent of Chimeric Antigen Receptor (CAR) T cell therapy, a new category of targetable biomarkers has emerged. These biomarkers are associated with the surface of malignant cells and serve as targets for directing cytotoxic T cells. The first biomarker target used for CAR T cell therapy was CD19, a B cell marker expressed highly on malignant B cells. With the success of CD19, the last decade has shown an explosion of new targetable biomarkers on a range of human malignancies. These surface targets have made it possible to provide directed, specific therapy that reduces healthy tissue destruction and preserves the patient's immune system during treatment. As of May 2018, there are over 100 clinical trials underway that target over 25 different surface biomarkers in almost every human tissue. This expansion has led to not only promising results in terms of patient outcome, but has also led to an exponential growth in the investigation of new biomarkers that could potentially be utilized in CAR T cell therapy for treating patients. In this review, we discuss the biomarkers currently under investigation and point out several promising biomarkers in the preclinical stage of development that may be useful as targets.
Collapse
Affiliation(s)
- Michelle H. Townsend
- Department of Microbiology and Molecular Biology, Brigham Young University, 3142 LSB, Provo, UT 84602 USA
| | - Gajendra Shrestha
- Department of Microbiology and Molecular Biology, Brigham Young University, 3142 LSB, Provo, UT 84602 USA
- Thunder Biotech, Highland, UT USA
| | - Richard A. Robison
- Department of Microbiology and Molecular Biology, Brigham Young University, 3142 LSB, Provo, UT 84602 USA
| | - Kim L. O’Neill
- Department of Microbiology and Molecular Biology, Brigham Young University, 3142 LSB, Provo, UT 84602 USA
| |
Collapse
|
8
|
Heinrich MC, Göbel C, Kluth M, Bernreuther C, Sauer C, Schroeder C, Möller-Koop C, Hube-Magg C, Lebok P, Burandt E, Sauter G, Simon R, Huland H, Graefen M, Heinzer H, Schlomm T, Heumann A. PSCA expression is associated with favorable tumor features and reduced PSA recurrence in operated prostate cancer. BMC Cancer 2018; 18:612. [PMID: 29855276 PMCID: PMC5984312 DOI: 10.1186/s12885-018-4547-7] [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: 10/11/2017] [Accepted: 05/22/2018] [Indexed: 11/28/2022] Open
Abstract
Background Prostate Stem Cell Antigen (PSCA) is frequently expressed in prostate cancer but its exact function is unclear. Methods To clarify contradictory findings on the prognostic role of PSCA expression, a tissue microarray containing 13,665 prostate cancers was analyzed by immunohistochemistry. Results PSCA staining was absent in normal epithelial and stromal cells of the prostate. Membranous and cytoplasmic PSCA staining was seen in 53.7% of 9642 interpretable tumors. Staining was weak in 22.4%, moderate in 24.5% and strong in 6.8% of tumors. PSCA expression was associated with favorable pathological and clinical tumor features: Early pathological tumor stage (p < 0.0001), low Gleason grade (p < 0.0001), absence of lymph node metastasis (p < 0.0001), low pre-operative PSA level (p = 0.0118), negative surgical margin (p < 0.0001) and reduced PSA recurrence (p < 0.0001). PSCA expression was an independent predictor of prognosis in multivariate analysis (hazard ratio 0.84, p < 0.0001). Conclusions The absence of statistical relationship to TMPRSS2:ERG fusion status, chromosomal deletion or high tumor cell proliferation argues against a major role of PSCA for regulation of cell cycle or genomic integrity. PSCA expression is linked to favorable prognosis. PSCA measurement is a candidate for inclusion in multi-parametric prognostic prostate cancer tests. Electronic supplementary material The online version of this article (10.1186/s12885-018-4547-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marie-Christine Heinrich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Cosima Göbel
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Christian Bernreuther
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Charlotte Sauer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Cornelia Schroeder
- Department of Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Christina Möller-Koop
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany.
| | - Hartwig Huland
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Markus Graefen
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Hans Heinzer
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Thorsten Schlomm
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany.,Department of Urology, Section for translational Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Asmus Heumann
- Department of Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| |
Collapse
|
9
|
Effect of Pycnogenol® on an experimental rat model of allergic conjunctivitis. Graefes Arch Clin Exp Ophthalmol 2018; 256:1299-1304. [DOI: 10.1007/s00417-018-3988-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/15/2018] [Accepted: 04/11/2018] [Indexed: 12/24/2022] Open
|
10
|
Kim SH, Park WS, Park BR, Joo J, Joung JY, Seo HK, Chung J, Lee KH. PSCA, Cox-2, and Ki-67 are independent, predictive markers of biochemical recurrence in clinically localized prostate cancer: a retrospective study. Asian J Androl 2018; 19:458-462. [PMID: 27232854 PMCID: PMC5507093 DOI: 10.4103/1008-682x.180798] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Prostate cancer is the second most common male cancer, with half of all patients going on to develop metastases. To better identify patients at high risk for prostate cancer progression and reduce prostate cancer-related mortality, improved prognostic factors are required. In this study, we used immunohistochemistry (IHC) to determine the prognostic values of multiple tissue biomarkers in hormone-naοve prostatectomy specimens of prostate cancer. Using 510 prostatectomy specimens collected between 2002 and 2012, IHC analysis was performed for Cerb-2, Cyclin D1, VEGF, EGFR, Rb, PSCA, p53, Bcl-2, Cox-2, PMS2, and Ki-67 on formalin-fixed paraffin-embedded sections. The Cox proportional hazard model was used to determine the predictive risk factors for biochemical recurrence (BCR) of prostate cancer. During a median 44-month follow-up, 128 (25.1%) patients developed BCR. A multivariate regression analysis revealed that Ki-67 (hazard ratio [HR]: 1.60, P = 0.033), PSCA (HR: 0.42, P < 0.001), and Cox-2 (HR: 2.05, P = 0.003) were the only significant prognostic tissue markers of BCR. Resection margin status (HR: 1.67, P = 0.010), pathologic pT0/1/2 stage (vs pT3/4; HR: 0.20, P = 0.002), preoperative PSA levels (HR: 1.03, P < 0.001), biopsied (HR: 1.30, P = 0.022) and pathologic (HR: 1.42, P = 0.005) Gleason scores, and prostate size (HR: 0.97, P = 0.003) were significant clinicopathologic factors. The expression of Ki-67, PSCA, and Cox-2 biomarkers along with other clinicopathologic factors were prognostic factors for BCR in patients with clinically localized prostate cancer following radical prostatectomy.
Collapse
Affiliation(s)
- Sung Han Kim
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Weon Seo Park
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea.,Department of Pathology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Bo Ram Park
- Department of Statistics, Biometric Research Branch, Clinical Research Coordination Center, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Jungnam Joo
- Department of Statistics, Biometric Research Branch, Clinical Research Coordination Center, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Jae Young Joung
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Ho Kyung Seo
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Jinsoo Chung
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Kang Hyun Lee
- Department of Urology, Center for Prostate Cancer, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| |
Collapse
|
11
|
Zettlitz KA, Tsai WTK, Knowles SM, Kobayashi N, Donahue TR, Reiter RE, Wu AM. Dual-Modality Immuno-PET and Near-Infrared Fluorescence Imaging of Pancreatic Cancer Using an Anti-Prostate Stem Cell Antigen Cys-Diabody. J Nucl Med 2018; 59:1398-1405. [PMID: 29602820 DOI: 10.2967/jnumed.117.207332] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/12/2018] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer has a high mortality rate due to late diagnosis and the tendency to invade surrounding tissues and metastasize at an early stage. A molecular imaging agent that enables both presurgery antigen-specific PET (immuno-PET) and intraoperative near-infrared fluorescence (NIRF) guidance might benefit diagnosis of pancreatic cancer, staging, and surgical resection, which remains the only curative treatment. Methods: We developed a dual-labeled probe based on A2 cys-diabody (A2cDb) targeting the cell-surface prostate stem cell antigen (PSCA), which is expressed in most pancreatic cancers. Maleimide-IRDye800CW was site-specifically conjugated to the C-terminal cys-tag (A2cDb-800) without impairing integrity or affinity (half-maximal binding, 4.3 nM). Direct radioiodination with 124I (124I-A2cDb-800) yielded a specific activity of 159 ± 48 MBq/mg with a radiochemical purity exceeding 99% and 65% ± 4.5% immunoreactivity (n = 3). In vivo specificity for PSCA-expressing tumor cells and biodistribution of the dual-modality tracer were evaluated in a prostate cancer xenograft model and compared with single-labeled 124I-A2cDb. Patient-derived pancreatic ductal adenocarcinoma xenografts (PDX-PDACs) were grown subcutaneously in NSG mice and screened for PSCA expression by immuno-PET. Small-animal PET/CT scans of PDX-PDAC-bearing mice were obtained using the dual-modality 124I-A2cDb-800 followed by postmortem NIRF imaging with the skin removed. Tumors and organs were analyzed ex vivo to compare the relative fluorescent signals without obstruction by other organs. Results: Specific uptake in PSCA-positive tumors and low nonspecific background activity resulted in high-contrast immuno-PET images. Concurrent with the PET studies, fluorescent signal was observed in the PSCA-positive tumors of mice injected with the dual-tracer 124I-A2cDb-800, with low background uptake or autofluorescence in the surrounding tissue. Ex vivo biodistribution confirmed comparable tumor uptake of both 124I-A2cDb-800 and 124I-A2cDb. Conclusion: Dual-modality imaging using the anti-PSCA cys-diabody resulted in high-contrast immuno-PET/NIRF images of PDX-PDACs, suggesting that this imaging agent might offer both noninvasive whole-body imaging to localize PSCA-positive pancreatic cancer and fluorescence image-guided identification of tumor margins during surgery.
Collapse
Affiliation(s)
- Kirstin A Zettlitz
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, California .,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.,David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Wen-Ting K Tsai
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, California.,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.,David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Scott M Knowles
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, California.,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.,David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Naoko Kobayashi
- David Geffen School of Medicine, UCLA, Los Angeles, California.,Department of Urology, UCLA, Los Angeles, California; and
| | - Timothy R Donahue
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.,David Geffen School of Medicine, UCLA, Los Angeles, California.,Division of General Surgery, Department of Surgery, UCLA, Los Angeles, California
| | - Robert E Reiter
- David Geffen School of Medicine, UCLA, Los Angeles, California.,Department of Urology, UCLA, Los Angeles, California; and
| | - Anna M Wu
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, California.,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.,David Geffen School of Medicine, UCLA, Los Angeles, California
| |
Collapse
|
12
|
Coskun A, Yavasoglu I, Meteoglu I, Unubol M, Yasar B, Borazan S, Omurlu IK, Yukselen V, Yasa MH. Vitamin D Receptor Level in Biopsy Specimen of Patients with Ulcerative Colitis: Results from a Center in Western Anatolia. J Natl Med Assoc 2017; 110:276-280. [PMID: 29778131 DOI: 10.1016/j.jnma.2017.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/09/2017] [Accepted: 06/09/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic, inflammatory bowel diseases characterized by uncontrolled inflammatory condition of the colon and rectal mucosa marked by recurrent periods of remission and exacerbation. Vitamin D receptor (VDR) is a member of the steroid receptor family that mediates the effects of vitamin D by regulating transcription of multiple cellular genes. We aimed to evaluate vitamin d receptor level in biopsy specimen of patients with UC in this study. METHODS VDR levels were retrospectively studied in colon biopsy specimens of UC patients. The Spearman's rho correlation analysis, The Kolmogorov-Smirnov, Mann Whitney U, and chi-square tests were used for statistical analysis. The p values below 0.05 were considered statistically significant. RESULTS Study included 112 UC patients (65 male and 47 female) and 30 controls (19 female and 11 male) who had normal results in biopsy examinations carried out due to various reasons. VDR levels of UC patients were statistically lower than control subjects, and was not associated with duration of the disease and place of involvement. CONCLUSIONS VDR is an important receptor in the pathogenesis of UC, and optimizing vitamin D levels could have a therapeutic role in UC.
Collapse
Affiliation(s)
- Adil Coskun
- Department of Gastroenterology, Adnan Menderes University, School of Medicine, TR-09100, Aydin, Turkey.
| | - Irfan Yavasoglu
- Department of Hematology, Adnan Menderes University, School of Medicine, TR-09100, Aydin, Turkey
| | - Ibrahim Meteoglu
- Department of Pathology, Adnan Menderes University, School of Medicine, TR-09100, Aydin, Turkey
| | - Mustafa Unubol
- Department of Endocrinology, Adnan Menderes University, School of Medicine, TR-09100, Aydin, Turkey
| | - Burak Yasar
- Department of Gastroenterology, Adnan Menderes University, School of Medicine, TR-09100, Aydin, Turkey
| | - Serkan Borazan
- Department of Gastroenterology, Adnan Menderes University, School of Medicine, TR-09100, Aydin, Turkey
| | - Imran Kurt Omurlu
- Department of Biostatistics, Adnan Menderes University, School of Medicine, TR-09100, Aydin, Turkey
| | - Vahit Yukselen
- Department of Gastroenterology, Adnan Menderes University, School of Medicine, TR-09100, Aydin, Turkey
| | - M Hadi Yasa
- Department of Gastroenterology, Adnan Menderes University, School of Medicine, TR-09100, Aydin, Turkey
| |
Collapse
|
13
|
Zhao Z, He J, Kang R, Zhao S, Liu L, Li F. RNA interference targeting PSCA suppresses primary tumor growth and metastasis formation of human prostate cancer xenografts in SCID mice. Prostate 2016; 76:184-98. [PMID: 26477693 DOI: 10.1002/pros.23110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 09/28/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVES Prostate stem cell antigen (PSCA) is a cell surface, glycosylphosphatidylinositol (GPI)-anchored glycoprotein. Its overexpression has been detected in both local and metastatic prostate cancer (PCa), making it a potential therapeutic target. We previously reported that silencing PSCA by small interfering RNA targeting human PSCA (siRNA-PSCA) inhibited biological activity of PSCA-positive PCa cells leading to reduced proliferation, motility and invasion in vitro. In this study, we extended this in vitro findings to in vivo settings in order to investigate the effects of this specific siRNA on the tumor growth and metastasis development of PCa in vivo. MATERIALS AND METHODS The siRNA-PSCA and ectopically overexpressed-PSCA vector were constructed and transfected into human PCa PC-3M and LNCaP cells, respectively, and were subcutaneously inoculated into the male SCID mice. Tumor growth was measured with a caliper, and formation of metastasis in mice bearing xenograft tumors was studied by magnetic resonance imaging (MRI) and autopsy analysis. Western blot and immunohistochemistry were used to assess the expression levels of PSCA protein in tumor tissues from xenograft and distant metastases. RESULTS Consistent with our previous in vitro findings, the subcutaneous xenografts of PC-3M-siPSCA exhibited the almost completely inhibited expression of PSCA protein in their tumors tissues (P < 0.001 and P < 0.001, respectively), and consequently had a significant reduction in tumor growth volumes (P < 0.05 for all), and metastasis onset and sites (P < 0.001 for all) compared to those of PC-3M and PC-3M-siScrm. Conversely, LNCaP-PSCA showed significantly enhanced primary tumor growth and metastasis formation of xenografts compared to LNCaP-vehicle and LNCaP cells (P < 0.001 for all). Moreover, the up-regulated expression of PSCA protein was detected in the distant metastases of xenograft tumors from all groups. CONCLUSIONS Taken together, these observations suggest that PSCA has a promoting role in the growth and metastasis of PCa and siRNA-PSCA may be a potential therapeutic strategy for PSCA-positive PCa.
Collapse
Affiliation(s)
- Zhigang Zhao
- Department of Urology and Andrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Jun He
- Department of Urology and Andrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Ran Kang
- Department of Urology and Andrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Shankun Zhao
- Department of Urology and Andrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Luhao Liu
- Department of Urology and Andrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Futian Li
- Department of Urology and Andrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
14
|
Esfahani M, Ataei N, Panjehpour M. Biomarkers for evaluation of prostate cancer prognosis. Asian Pac J Cancer Prev 2016; 16:2601-11. [PMID: 25854335 DOI: 10.7314/apjcp.2015.16.7.2601] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Prostate cancer, with a lifetime prevalence of one in six men, is the second cause of malignancy-related death and the most prevalent cancer in men in many countries. Nowadays, prostate cancer diagnosis is often based on the use of biomarkers, especially prostate-specific antigen (PSA) which can result in enhanced detection at earlier stage and decreasing in the number of metastatic patients. However, because of the low specificity of PSA, unnecessary biopsies and mistaken diagnoses frequently occur. Prostate cancer has various features so prognosis following diagnosis is greatly variable. There is a requirement for new prognostic biomarkers, particularly to differentiate between inactive and aggressive forms of disease, to improve clinical management of prostate cancer. Research continues into finding additional markers that may allow this goal to be attained. We here selected a group of candidate biomarkers including PSA, PSA velocity, percentage free PSA, TGFβ1, AMACR, chromogranin A, IL-6, IGFBPs, PSCA, biomarkers related to cell cycle regulation, apoptosis, PTEN, androgen receptor, cellular adhesion and angiogenesis, and also prognostic biomarkers with Genomic tests for discussion. This provides an outline of biomarkers that are presently of prognostic interest in prostate cancer investigation.
Collapse
Affiliation(s)
- Maryam Esfahani
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran E-mail :
| | | | | |
Collapse
|
15
|
Nuclear Factor Kappa B, Matrix Metalloproteinase-1, p53, and Ki-67 Expressions in the Primary Tumors and the Lymph Node Metastases of Colorectal Cancer Cases. Gastroenterol Res Pract 2015; 2015:945392. [PMID: 25945089 PMCID: PMC4402505 DOI: 10.1155/2015/945392] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 03/20/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most frequent malignancy. Many factors such as NF-κB, matrix metalloproteinase-1 (MMP-1), p53, and Ki-67 are likely to be involved in its development and progression. Lymph node metastases indicate increased tumor burden and tumor cell heterogeneity and affect both the treatment strategies and the prognosis. In this study, expressions of NF-κB, MMP-1, p53, and Ki-67 were between the primary tumors and lymph node metastases in 110 Dukes' stage C, CRC cases by immunohistochemical methods, related to patients' clinical outcomes. NF-κB, p53, and Ki-67 expressions were significantly higher in the metastatic lymph nodes compared to the primary tumor tissues (P = 0.04, P = 0.04, and P = 0.01, resp.). In the metastatic lymph nodes NF-κB expression was correlated with both p53 (r = 0.546, P = 0.003) and Ki-67 (r = 0.586, P = 0.0001) expressions. The univariant and multivariant analyses showed that only “pT stage” preserved an independent prognostic significance for recurrence-free survival rates and 5-year overall survival rates (P < 0.001 for both). Metastatic cells can acquire different biological characteristics compared to their primaries. Elucidation of properties acquired by metastatic cells is important in order to better determine prognosis, reverse drug resistance, and discover new treatment alternatives.
Collapse
|
16
|
Assi J, Srivastava G, Matta A, MacMillan C, Ralhan R, Walfish PG. Nuclear Ep-ICD expression is a predictor of poor prognosis in "low risk" prostate adenocarcinomas. PLoS One 2015; 10:e0107586. [PMID: 25695234 PMCID: PMC4335027 DOI: 10.1371/journal.pone.0107586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 08/12/2014] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Molecular markers for predicting prostate cancer (PCa) that would have poor prognosis are urgently needed for a more personalized treatment for patients. Regulated intramembrane proteolysis of Epithelial cell adhesion molecule results in shedding of the extracellular domain (EpEx) and release of its intracellular domain (Ep-ICD) which triggers oncogenic signaling and might correlate to tumor aggressiveness. This study aimed to explore the potential of Ep-ICD and EpEx to identify PCa that have poor prognosis. METHODS Immunohistochemical analysis of Ep-ICD and EpEx was carried out in normal prostate tissues (n = 100), benign prostate hyperplasia (BPH, n = 83), and prostate cancer (n = 249) using domain specific antibodies. The expression of Ep-ICD and EpEx was correlated with clinico- pathological parameters and disease free survival (DFS). RESULTS Reduced expression of nuclear Ep-ICD and membrane EpEx was observed in PCa in comparison with BPH and normal prostate tissues (p = 0.006, p < 0.001 respectively). For patients who had PCa with Gleason Score less than 7, preserved nuclear Ep-ICD emerged as the most significant marker in multivariate analysis for prolonged DFS, where these patients did not have recurrence during follow up of up to 12 years (p = 0.001). CONCLUSION Reduced expression of nuclear Ep-ICD was associated with shorter disease free survival in patients with a Gleason Score less than 7 and may be useful in identifying patients likely to have aggressive tumors with poor prognosis. Furthermore, nuclear Ep-ICD can differentiate between normal and prostate cancer tissues for ambiguous cases.
Collapse
Affiliation(s)
- Jasmeet Assi
- Alex and Simona Shnaider Laboratory in Molecular Oncology, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, Canada
| | - Gunjan Srivastava
- Alex and Simona Shnaider Laboratory in Molecular Oncology, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, Canada
| | - Ajay Matta
- Alex and Simona Shnaider Laboratory in Molecular Oncology, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, Canada
| | - Christina MacMillan
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ranju Ralhan
- Alex and Simona Shnaider Laboratory in Molecular Oncology, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Otolaryngology—Head and Neck Surgery, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Otolaryngology—Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- * E-mail: (PGW); (RR)
| | - Paul G. Walfish
- Alex and Simona Shnaider Laboratory in Molecular Oncology, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Medicine, Endocrine Division, Mount Sinai Hospital and University of Toronto, Toronto, Ontario, Canada
- Department of Otolaryngology—Head and Neck Surgery, Mount Sinai Hospital, Toronto, Ontario, Canada
- * E-mail: (PGW); (RR)
| |
Collapse
|
17
|
Knowles SM, Tavaré R, Zettlitz KA, Rochefort MM, Salazar FB, Jiang ZK, Reiter RE, Wu AM. Applications of immunoPET: using 124I-anti-PSCA A11 minibody for imaging disease progression and response to therapy in mouse xenograft models of prostate cancer. Clin Cancer Res 2014; 20:6367-78. [PMID: 25326233 DOI: 10.1158/1078-0432.ccr-14-1452] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Prostate stem cell antigen (PSCA) is highly expressed in local prostate cancers and prostate cancer bone metastases and its expression correlates with androgen receptor activation and a poor prognosis. In this study, we investigate the potential clinical applications of immunoPET with the anti-PSCA A11 minibody, an antibody fragment optimized for use as an imaging agent. We compare A11 minibody immunoPET to (18)F-Fluoride PET bone scans for detecting prostate cancer bone tumors and evaluate the ability of the A11 minibody to image tumor response to androgen deprivation. EXPERIMENTAL DESIGN Osteoblastic, PSCA-expressing, LAPC-9 intratibial xenografts were imaged with serial (124)I-anti-PSCA A11 minibody immunoPET and (18)F-Fluoride bone scans. Mice bearing LAPC-9 subcutaneous xenografts were treated with either vehicle or MDV-3100 and imaged with A11 minibody immunoPET/CT scans pre- and posttreatment. Ex vivo flow cytometry measured the change in PSCA expression in response to androgen deprivation. RESULTS A11 minibody demonstrated improved sensitivity and specificity over (18)F-Fluoride bone scans for detecting LAPC-9 intratibial xenografts at all time points. LAPC-9 subcutaneous xenografts showed downregulation of PSCA when treated with MDV-3100 which A11 minibody immunoPET was able to detect in vivo. CONCLUSIONS A11 minibody immunoPET has the potential to improve the sensitivity and specificity of clinical prostate cancer metastasis detection over bone scans, which are the current clinical standard-of-care. A11 minibody immunoPET additionally has the potential to image the activity of the androgen signaling axis in vivo which may help evaluate the clinical response to androgen deprivation and the development of castration resistance.
Collapse
Affiliation(s)
- Scott M Knowles
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California
| | - Richard Tavaré
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California
| | - Kirstin A Zettlitz
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California
| | - Matthew M Rochefort
- Department of Surgery, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California
| | - Felix B Salazar
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California
| | - Ziyue Karen Jiang
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California. Department of Urology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California
| | - Robert E Reiter
- Department of Urology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California
| | - Anna M Wu
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California.
| |
Collapse
|
18
|
Barve A, Jin W, Cheng K. Prostate cancer relevant antigens and enzymes for targeted drug delivery. J Control Release 2014; 187:118-32. [PMID: 24878184 DOI: 10.1016/j.jconrel.2014.05.035] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 05/13/2014] [Accepted: 05/17/2014] [Indexed: 12/26/2022]
Abstract
Chemotherapy is one of the most widely used approaches in combating advanced prostate cancer, but its therapeutic efficacy is usually insufficient due to poor specificity and associated toxicity. Lack of targeted delivery to prostate cancer cells is also the primary obstacles in achieving feasible therapeutic effect of other promising agents including peptide, protein, and nucleic acid. Consequently, there remains a critical need for strategies to increase the selectivity of anti-prostate cancer agents. This review will focus on various prostate cancer-relevant antigens and enzymes that could be exploited for prostate cancer targeted drug delivery. Among various targeting strategies, active targeting is the most advanced approach to specifically deliver drugs to their designated cancer cells. In this approach, drug carriers are modified with targeting ligands that can specifically bind to prostate cancer-specific antigens. Moreover, there are several specific enzymes in the tumor microenvironment of prostate cancer that can be exploited for stimulus-responsive drug delivery systems. These systems can specifically release the active drug in the tumor microenvironment of prostate cancer, leading to enhanced tumor penetration efficiency.
Collapse
Affiliation(s)
- Ashutosh Barve
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City 64108, USA
| | - Wei Jin
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City 64108, USA
| | - Kun Cheng
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City 64108, USA.
| |
Collapse
|
19
|
Novel tools for prostate cancer prognosis, diagnosis, and follow-up. BIOMED RESEARCH INTERNATIONAL 2014; 2014:890697. [PMID: 24877145 PMCID: PMC4024423 DOI: 10.1155/2014/890697] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 04/09/2014] [Indexed: 12/18/2022]
Abstract
Prostate-specific antigen (PSA) is the main diagnostic tool when it comes to prostate cancer but it possesses serious limitations. Therefore, there is an urgent need for more sensitive and specific biomarkers for prostate cancer prognosis and patient follow-up. Recent advances led to the discovery of many novel diagnostic/prognostic techniques and provided us with many worthwhile candidates. This paper briefly reviews the most promising biomarkers with respect to their implementation in screening, early detection, diagnostic confirmation, prognosis, and prediction of therapeutic response or monitoring disease and recurrence; and their use as possible therapeutic targets. This review also examines the possible future directions in the field of prostate cancer marker research.
Collapse
|
20
|
Knowles SM, Zettlitz KA, Tavaré R, Rochefort MM, Salazar FB, Stout DB, Yazaki PJ, Reiter RE, Wu AM. Quantitative immunoPET of prostate cancer xenografts with 89Zr- and 124I-labeled anti-PSCA A11 minibody. J Nucl Med 2014; 55:452-9. [PMID: 24504052 DOI: 10.2967/jnumed.113.120873] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
UNLABELLED Prostate stem cell antigen (PSCA) is expressed on the cell surface in 83%-100% of local prostate cancers and 87%-100% of prostate cancer bone metastases. In this study, we sought to develop immunoPET agents using (124)I- and (89)Zr-labeled anti-PSCA A11 minibodies (scFv-CH3 dimer, 80 kDa) and evaluate their use for quantitative immunoPET imaging of prostate cancer. METHODS A11 anti-PSCA minibody was alternatively labeled with (124)I- or (89)Zr-desferrioxamine and injected into mice bearing either matched 22Rv1 and 22Rv1×PSCA or LAPC-9 xenografts. Small-animal PET data were obtained and quantitated with and without recovery coefficient-based partial-volume correction, and the results were compared with ex vivo biodistribution. RESULTS Rapid and specific localization to PSCA-positive tumors and high-contrast imaging were observed with both (124)I- and (89)Zr-labeled A11 anti-PSCA minibody. However, the differences in tumor uptake and background uptake of the radiotracers resulted in different levels of imaging contrast. The nonresidualizing (124)I-labeled minibody had lower tumor uptake (3.62 ± 1.18 percentage injected dose per gram [%ID/g] 22Rv1×PSCA, 3.63 ± 0.59 %ID/g LAPC-9) than the residualizing (89)Zr-labeled minibody (7.87 ± 0.52 %ID/g 22Rv1×PSCA, 9.33 ± 0.87 %ID/g LAPC-9, P < 0.0001 for each), but the (124)I-labeled minibody achieved higher imaging contrast because of lower nonspecific uptake and better tumor-to-soft-tissue ratios (22Rv1×PSCA:22Rv1 positive-to-negative tumor, 13.31 ± 5.59 (124)I-A11 and 4.87 ± 0.52 (89)Zr-A11, P = 0.02). Partial-volume correction was found to greatly improve the correspondence between small-animal PET and ex vivo quantification of tumor uptake for immunoPET imaging with both radionuclides. CONCLUSION Both (124)I- and (89)Zr-labeled A11 anti-PSCA minibody showed high-contrast imaging of PSCA expression in vivo. However, the (124)I-labeled A11 minibody was found to be the superior imaging agent because of lower nonspecific uptake and higher tumor-to-soft-tissue contrast. Partial-volume correction was found to be essential for robust quantification of immunoPET imaging with both (124)I- and (89)Zr-labeled A11 minibody.
Collapse
Affiliation(s)
- Scott M Knowles
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bradley JL, Edwards CS, Fullard RJ. Adaptation of impression cytology to enable conjunctival surface cell transcriptome analysis. Curr Eye Res 2013; 39:31-41. [PMID: 24047118 DOI: 10.3109/02713683.2013.823213] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE This study investigates the extent of the human transcriptome that can be quantified from conjunctival impression cytology extracts. The aim is to determine if sufficient RNA can be isolated from a patient's conjunctival surface to identify differences in gene expression between dry eye and normal patients of (a) an array of 96 inflammatory biomarkers and associated receptors, and (b) if this comparison can be expanded to the entire transcriptome. MATERIALS AND METHODS CIC was used to collect conjunctival surface cells from 53 qualifying normal and dry eye patients. Based on prior optimization of all assay steps, RNA was isolated from the samples using a Qiagen RNeasy Plus Mini Kit and qRT-PCR was used to determine gene expression of 96 genes using TaqMan Low Density Array cards. Samples from six normal and six dry eye patients were then assayed on an Illumina Human HT-12 BeadChip. RESULTS Optimization steps yielded an RNA processing procedure that improved yield from an initial 12 genes through 96, then to the entire human transcriptome. For the HT-12 BeadChip, more than 30 genes differed by a factor of >1.5 between the dry eye and normal groups and seven genes were down-regulated by a factor of >2.0 in the dry eye group: HLA-DRB5, PSCA, FOS, lysozyme, TSC22D1, CAPN13 and CXCL6. CONCLUSIONS Conjunctival impression cytology can be used to collect sufficient RNA from conjunctival surface cells that, when processed optimally, allows successful transcriptome-wide expression analysis. While the current transcriptome analysis used a limited patient group, larger studies of patients with various types and severities of dry eye should reveal significant gene expression trends that can then be targeted to improve dry eye treatment options.
Collapse
Affiliation(s)
- John L Bradley
- School of Optometry, University of Alabama at Birmingham , Birmingham, AL , USA
| | | | | |
Collapse
|
22
|
Abstract
Prostate cancer (PCa), a highly heterogeneous disease, is the one of the leading cause of morbidity and mortality in the developed countries. Historically used biomarkers such as prostatic acid phosphatase (PAP), serum prostate-specific antigen (PSA), and its precursor have not stood the challenge of sensitivity and specificity. At present, there is need to re-evaluate the approach to diagnose and monitor PCa. To this end, molecular markers that can accurately identify men with PCa at an early stage, and those who would benefit from early therapeutic intervention, are the need of the hour. There has been unprecedented progress in the development of new PCa biomarkers through advancements in proteomics, tissue DNA and protein/RNA microarray, identification of microRNA, isolation of circulating tumor cells, and tumor immunohistochemistry. This review will examine the current status of prostate cancer biomarkers with emphasis on emerging biomarkers by evaluating their diagnostic and prognostic potentials.
Collapse
Affiliation(s)
- Tapan Bhavsar
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | |
Collapse
|
23
|
Fang CY, Huang SY, Wu CC, Hsu HY, Chou SP, Tsai CH, Chang Y, Takada K, Chen JY. The synergistic effect of chemical carcinogens enhances Epstein-Barr virus reactivation and tumor progression of nasopharyngeal carcinoma cells. PLoS One 2012; 7:e44810. [PMID: 23024765 PMCID: PMC3443098 DOI: 10.1371/journal.pone.0044810] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 08/07/2012] [Indexed: 12/04/2022] Open
Abstract
Seroepidemiological studies imply a correlation between Epstein-Barr virus (EBV) reactivation and the development of nasopharyngeal carcinoma (NPC). N-nitroso compounds, phorbols, and butyrates are chemicals found in food and herb samples collected from NPC high-risk areas. These chemicals have been reported to be risk factors contributing to the development of NPC, however, the underlying mechanism is not fully understood. We have demonstrated previously that low dose N-methyl-N’-nitro-N-nitrosoguanidine (MNNG, 0.1 µg/ml) had a synergistic effect with 12-O-tetradecanoylphorbol-13-acetate (TPA) and sodium butyrate (SB) in enhancing EBV reactivation and genome instability in NPC cells harboring EBV. Considering that residents in NPC high-risk areas may contact regularly with these chemical carcinogens, it is vital to elucidate the relation between chemicals and EBV and their contributions to the carcinogenesis of NPC. In this study, we constructed a cell culture model to show that genome instability, alterations of cancer hallmark gene expression, and tumorigenicity were increased after recurrent EBV reactivation in NPC cells following combined treatment of TPA/SB and MNNG. NPC cells latently infected with EBV, NA, and the corresponding EBV-negative cell, NPC-TW01, were periodically treated with MNNG, TPA/SB, or TPA/SB combined with MNNG. With chemically-induced recurrent reactivation of EBV, the degree of genome instability was significantly enhanced in NA cells treated with a combination of TPA/SB and MNNG than those treated individually. The Matrigel invasiveness, as well as the tumorigenicity in mouse, was also enhanced in NA cells after recurrent EBV reactivation. Expression profile analysis by microarray indicates that many carcinogenesis-related genes were altered after recurrent EBV reactivation, and several aberrations observed in cell lines correspond to alterations in NPC lesions. These results indicate that cooperation between chemical carcinogens can enhance the reactivation of EBV and, over recurrent reactivations, lead to alteration of cancer hallmark gene expression with resultant enhancement of tumorigenesis in NPC.
Collapse
Affiliation(s)
- Chih-Yeu Fang
- National Institute of Cancer Research, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Sheng-Yen Huang
- Graduate Program of Biotechnology in Medicine of National Tsing Hua University and National Health Research Institutes, Hsinchu, Taiwan
- Institute of Biotechnology, Department of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chung-Chun Wu
- National Institute of Cancer Research, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Hui-Yu Hsu
- Institute of Biotechnology, Department of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Sheng-Ping Chou
- National Institute of Cancer Research, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Ching-Hwa Tsai
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yao Chang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| | - Kenzo Takada
- Department of Tumor Virology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Jen-Yang Chen
- National Institute of Cancer Research, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
24
|
Wang T, Zhang L, Li H, Wang B, Chen K. Prostate stem cell antigen polymorphisms and susceptibility to gastric cancer: a systematic review and meta-analysis. Cancer Epidemiol Biomarkers Prev 2012; 21:843-50. [PMID: 22426141 DOI: 10.1158/1055-9965.epi-11-1176] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Many studies have reported that prostate stem cell antigen (PSCA) polymorphisms (rs2294008 and/or 2976392) are significantly associated with gastric cancer (GC) risk, although the published results are inconsistent. We conducted a systematic review and meta-analysis for relevant literatures to quantitatively evaluate the relationship between PSCA polymorphisms and GC susceptibility. METHODS Extensive searches were conducted in three databases up to November 1, 2011. ORs and 95% CIs were used to assess the strength of the associations. The data were further stratified by ethnicity, histopathology, subsite, and study design. All of the associations were evaluated with dominant model and recessive model, respectively. Heterogeneity and publication bias were also assessed by Q test, I(2), and funnel plot accordingly. RESULTS Nine articles including 11 case-control data sets were included, with 10,746 GC cases and 9,158 controls for rs2294008 and 6,060 cases and 4,824 controls for rs2976392. The results showed that risk allele carriers were significantly associated with GC risk compared with nonrisk allele homozygotes. In stratification analyses, these associations remained significant for majority of subgroups except for Caucasians and noncardia tumor in dominant model, and cardia tumor in both dominant and recessive model. Random model was used when heterogeneity among studies was detected. No publication bias was observed. CONCLUSIONS The two loci of PSCA (rs2294008 and rs2976392) were both significantly associated with GC susceptibility and in linkage disequilibrium. IMPACT More prospective studies on PSCA polymorphisms at multicenters with sufficient sample size and less heterogeneity will be needed for further validations.
Collapse
Affiliation(s)
- Tao Wang
- Department of Epidemiology and Biostatistics, Tianjin Medical University, Tianjin, China
| | | | | | | | | |
Collapse
|
25
|
Karan D, Dubey S, Van Veldhuizen P, Holzbeierlein JM, Tawfik O, Thrasher JB. Dual antigen target-based immunotherapy for prostate cancer eliminates the growth of established tumors in mice. Immunotherapy 2012; 3:735-46. [PMID: 21668311 DOI: 10.2217/imt.11.59] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
AIMS We have previously shown that immunization with an adenovirus vector carrying an individual antigen induces antigen-specific CD8 T cells actively engaged in the destruction of tumor cells expressing the cognate antigen. In order to expand the range of antitumor responses beyond an individual antigen, we designed a recombinant adenovirus type 5 (rAd5) carrying a fusion construct of two full-length antigens. We used this adenovirus vector to test the concept that multiantigenic effector T cells could be generated simultaneously following a single immunization. METHOD To perform the rAd5 constructs, we selected a combination of prostate-specific antigen (PSA) and prostate stem cell antigen (PSCA) genes based on their restricted distribution within the prostate tissue and their association with the development and progression of prostate cancer. RESULTS Immunization of mice with rAd5 vector carrying a fusion construct of PSA and PSCA (Ad5-PSA/PSCA) simultaneously induced the expansion of anti-PSA and anti-PSCA CD8 T cells, as measured by intracellular cytokine staining for IFN-γ. The antigen-specific T-cell responses that developed were efficient in eliminating the target cells expressing cognate antigens measured by an in vivo cytotoxic T-cell assay. The in vivo tumor growth study showed that immunization of mice with Ad5-PSA/PSCA vaccine induced strong antitumor immunity when challenged with mouse prostate tumor cell lines (RM11) expressing human PSA (RM11/PSA). To further analyze the impact on therapeutic efficacy of Ad5-PSA/PSCA vaccine against the tumor cells expressing PSA and PSCA (RM11-PSA/PSCA) antigens, we injected mice with Ad5-PSA/PSCA vaccine. The vaccine inhibited the growth of established tumors with 80% of the mice becoming tumor free. These data provide useful information that antigen-specific effector T cells can be generated simultaneously and that their additive antitumor effect has the potential to eliminate the growth of established tumors. Therefore, the immunotherapy approach of using the simultaneous targeting of dual antigens associated with prostate cancer may have important implications for human clinical trials.
Collapse
Affiliation(s)
- Dev Karan
- Veterans Affairs Medical Center, Kansas City, MO, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Larkin SET, Holmes S, Cree IA, Walker T, Basketter V, Bickers B, Harris S, Garbis SD, Townsend PA, Aukim-Hastie C. Identification of markers of prostate cancer progression using candidate gene expression. Br J Cancer 2011; 106:157-65. [PMID: 22075945 PMCID: PMC3251845 DOI: 10.1038/bjc.2011.490] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Metastatic prostate cancer (PCa) has no curative treatment options. Some forms of PCa are indolent and slow growing, while others metastasise quickly and may prove fatal within a very short time. The basis of this variable prognosis is poorly understood, despite considerable research. The aim of this study was to identify markers associated with the progression of PCa. METHODS Artificial neuronal network analysis combined with data from literature and previous work produced a panel of putative PCa progression markers, which were used in a transcriptomic analysis of 29 radical prostatectomy samples and correlated with clinical outcome. RESULTS Statistical analysis yielded seven putative markers of PCa progression, ANPEP, ABL1, PSCA, EFNA1, HSPB1, INMT and TRIP13. Two data transformation methods were utilised with only markers that were significant in both selected for further analysis. ANPEP and EFNA1 were significantly correlated with Gleason score. Models of progression co-utilising markers ANPEP and ABL1 or ANPEP and PSCA had the ability to correctly predict indolent or aggressive disease, based on Gleason score, in 89.7% and 86.2% of cases, respectively. Another model of TRIP13 expression in combination with preoperative PSA level and Gleason score was able to correctly predict recurrence in 85.7% of cases. CONCLUSION This proof of principle study demonstrates a novel association of carcinogenic and tumourigenic gene expression with PCa stage and prognosis.
Collapse
Affiliation(s)
- S E T Larkin
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St Michaels Building, White Swan Road, Portsmouth, PO1 2DT, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhao Z, Ma W, Zeng G, Qi D, Ou L, Liang Y. Small interference RNA-mediated silencing of prostate stem cell antigen attenuates growth, reduces migration and invasion of human prostate cancer PC-3M cells. Urol Oncol 2011; 31:343-51. [PMID: 21429770 DOI: 10.1016/j.urolonc.2011.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Revised: 02/04/2011] [Accepted: 02/07/2011] [Indexed: 12/01/2022]
Abstract
OBJECTIVES Prostate stem cell antigen (PSCA), a glycosylphosphatidylinositol (GPI)-anchored cell surface glycoprotein, is highly expressed in both local and metastatic prostate cancer (CaP). Elevated PSCA expression has been shown to correlate with malignant phenotype and clinical progression. The purpose of the current study is to investigate the therapeutic potential of small interference RNA (siRNA) targeting PSCA on human CaP cells. MATERIALS AND METHODS A set of two siRNAs directed different regions of human PSCA (siRNA-PSCA) were designed and transfected into a human CaP PC-3M cell line. The silencing effect was screened by RT-PCR and Western blotting. The biological effects of siRNA-PSCA on PC-3M cells were investigated by examining the cell proliferation through 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, cell cycle distribution through flow cytometry, and migration and invasion potencies through transwell invasion assay upon the PSCA silencing. RESULTS PC-3M cells had positive PSCA expression on immunocytochemical assay. PSCA expression was depleted at 48 hours after transfection with siRNA-PSCA. Silencing of PSCA significantly suppressed cell proliferation. Cell cycle assay showed that the anti-proliferation effect of siRNA-PSCA was mediated by arresting cells in the G0/G1 phase rather than apoptosis. Furthermore, PSCA knockdown resulted in a marked decrease of cell migration and invasion capabilities in PC-3M cells. CONCLUSIONS The present study provides the first evidence that silencing PSCA using siRNA can inhibit the proliferation and invasiveness properties of human CaP cells, which may provide a promising therapeutic strategy for CaP and open a novel avenue toward the investigation of the role of PSCA overexpression in cancers.
Collapse
Affiliation(s)
- Zhigang Zhao
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical College, Guangdong Key Laboratory of Urology, Guangzhou, China.
| | | | | | | | | | | |
Collapse
|
28
|
Li Y, Cozzi PJ, Russell PJ. Promising tumor-associated antigens for future prostate cancer therapy. Med Res Rev 2010; 30:67-101. [PMID: 19536865 DOI: 10.1002/med.20165] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Prostate cancer (CaP) is one of the most prevalent malignant diseases among men in Western countries. There is currently no cure for metastatic castrate-resistant CaP, and median survival for these patients is about 18 months; the high mortality rate seen is associated with widespread metastases. Progression of CaP from primary to metastatic disease is associated with several molecular and genetic changes that can affect the expression of specific tumor-associated antigens (TAAs) or receptors on the cell surface. Targeting TAAs is emerging as an area of promise for controlling late-stage and recurrent CaP. Several reviews have summarized the progress made in targeting signaling pathways for CaP but will not be discussed here. We describe some important CaP TAAs. These include prostate stem-cell antigen, prostate-specific membrane antigen, MUC1, epidermal growth factor receptor, platelet-derived growth factor and its receptor, urokinase plasminogen activator and its receptor, and extracellular matrix metalloproteinase inducer. We summarize recent advancements in our understanding of their role in CaP metastasis, as well as potential therapeutic options for targeting CaP TAAs. We also discuss the origin, identification, and characterization of prostate cancer stem cells (CSCs) and the potential benefits of targeting prostate CSCs to overcome chemoresistance and CaP recurrence.
Collapse
Affiliation(s)
- Yong Li
- Cancer Care Centre, St. George Hospital, Sydney, NSW, Australia.
| | | | | |
Collapse
|
29
|
Zhao Z, Liu J, Li S, Shen W. Prostate stem cell antigen mRNA expression in preoperatively negative biopsy specimens predicts subsequent cancer after transurethral resection of the prostate for benign prostatic hyperplasia. Prostate 2009; 69:1292-302. [PMID: 19462463 DOI: 10.1002/pros.20973] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Recent data showed that prostate stem cell antigen (PSCA) mRNA expression in transurethral resection of the prostate (TURP)-resected tissues predicted the subsequent prostate cancer after TURP in benign prostatic hyperplasia (BPH) patients with both PSA < 4.0 ng/ml and normal DRE findings. This study was to determine whether PSCA mRNA positivity in preoperatively negative prostatic biopsy samples from BPH men with PSA > 4.0 ng/ml and/or suspicious DRE findings had predictive performance following TURP. MATERIALS AND METHODS PSCA in situ hybridization was performed on negative prostatic biopsies taken before TURP from 166 enrolled symptomatic BPH patients, who were continuously followed for 5 years postoperatively. Predictive performance of PSCA mRNA for subsequent cancer onset was evaluated by univariate and multivariate Cox proportional hazards models with bootstrapping and concordance indices. RESULTS PSCA mRNA was detected in 42/166 (25.3%) of the preoperatively negative biopsy specimens, with a mean positive-labeling cells of 31.6%, in which 31 patients were identified as having subsequent PCa on follow-up. Of 124 patients with negative expression for PSCA mRNA none were subsequently diagnosed with PCa. The examination of Spearman's rank correlation coefficient showed that PSCA mRNA expression levels were positively and statistically correlated with higher Gleason score (r = 0.88, P < 0.001) and clinical T stage (r = 0.84, P < 0.001). A final multivariate Cox proportional hazards model demonstrated that only PSCA mRNA expression in negative prostatic biopsies was predictive of the subsequent cancer development after TURP (hazard ratio = 3.49; 95% CI: 2.02-4.75; P < 0.001), with the concordance index of 0.893. CONCLUSIONS This prospective study identifies PSCA mRNA in preoperatively negative prostatic biopsies as a significant predictor of subsequent cancer after TURP.
Collapse
Affiliation(s)
- Zhigang Zhao
- Department of Urology, The Second Affiliated Hospital, Shantou University Medical College, Guangdong Province, China.
| | | | | | | |
Collapse
|
30
|
Minimum information specification for in situ hybridization and immunohistochemistry experiments (MISFISHIE). Nat Biotechnol 2008; 26:305-12. [PMID: 18327244 DOI: 10.1038/nbt1391] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
One purpose of the biomedical literature is to report results in sufficient detail that the methods of data collection and analysis can be independently replicated and verified. Here we present reporting guidelines for gene expression localization experiments: the minimum information specification for in situ hybridization and immunohistochemistry experiments (MISFISHIE). MISFISHIE is modeled after the Minimum Information About a Microarray Experiment (MIAME) specification for microarray experiments. Both guidelines define what information should be reported without dictating a format for encoding that information. MISFISHIE describes six types of information to be provided for each experiment: experimental design, biomaterials and treatments, reporters, staining, imaging data and image characterizations. This specification has benefited the consortium within which it was developed and is expected to benefit the wider research community. We welcome feedback from the scientific community to help improve our proposal.
Collapse
|
31
|
Siva AC, Wild MA, Kirkland RE, Nolan MJ, Lin B, Maruyama T, Yantiri-Wernimont F, Frederickson S, Bowdish KS, Xin H. Targeting CUB domain-containing protein 1 with a monoclonal antibody inhibits metastasis in a prostate cancer model. Cancer Res 2008; 68:3759-66. [PMID: 18483259 DOI: 10.1158/0008-5472.can-07-1657] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Through a whole-cell panning approach, we previously identified a panel of antibodies that bound to prostate cancer cell surface antigens. One such antigen, CUB domain-containing protein 1 (CDCP1), was recognized by monoclonal antibody 25A11 and is a single transmembrane molecule highly expressed in several metastatic cancers as well as on CD34(+)CD133(+) myeloid leukemic blast cells. We show CDCP1 expression on prostate cancer cell lines by real-time quantitative PCR (RT-qPCR), flow cytometry, and immunohistochemistry and on prostate cancer patient samples by RT-qPCR and immunohistochemical staining. In cell-based assays, antibody 25A11 inhibited prostate cancer cell migration and invasion in vitro. Further characterization showed that CDCP1 is internalized on antibody binding. When 25A11 was coupled to the cytotoxin saporin either directly or via a secondary antibody, both resulted in prostate cancer cell killing in vitro. In vivo targeting studies with an anti-CDCP1 immunotoxin showed significant inhibition of primary tumor growth as well as metastasis in a mouse xenograft model. These data provide support for continued evaluation of anti-CDCP1 therapy for potential use in cancer in primary and metastatic disease.
Collapse
Affiliation(s)
- Amara C Siva
- Alexion Antibody Technologies, Inc., San Diego, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Halvorsen OJ. Molecular and prognostic markers in prostate cancer. APMIS 2008. [DOI: 10.1111/j.1600-0463.2008.0s123.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
33
|
Zhigang Z, Wenlu S. Flutamide reduced prostate cancer development and prostate stem cell antigen mRNA expression in high grade prostatic intraepithelial neoplasia. Int J Cancer 2008; 122:864-70. [PMID: 17957793 DOI: 10.1002/ijc.23150] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
High-grade prostatic intraepithelial neoplasia (HGPIN) appears to represent an ideal target for chemoprevention of prostate cancer (PCa). HGPIN responds to androgen ablation and has prostate stem cell antigen (PSCA) mRNA expression. One hundred and seventy two patients with isolated HGPIN were randomized in a double-blind manner to receive flutamide 250 mg/day (86 cases) or a placebo (86 cases) for 12 months and were rebiopsied at 12 and 60 months. PSCA mRNA expression was assessed in the prestudy and 12-month biopsies by in situ hybridization. The incidence of subsequent PCa was 11.6% in the flutamide group when compared with 30.2% in the placebo group over a follow-up period of 5 years (p = 0.0027). PSCA mRNA expression levels were significantly declined after treatment compared with that before treatment (p < 0.001). After treatment, 66 patients had reduced PSCA mRNA expression, in whom none was found with cancer on follow-up, however, 13 cases had increased PSCA mRNA expression levels, in whom 11 were found with cancer. Cox regression analysis demonstrated that HGPIN with increased PSCA mRNA expression after flutamide had an increased relative risk of 4.33 to develop subsequent cancer (95% confidence intervals: 2.48-7.36; p < 0.001). Seventeen (19.8%) cases had the flutamide-associated side effects, which were graded as mild, but all did not discontinue study. Flutamide can effectively and safely reduce PCa development and significantly suppress PSCA mRNA expression in men with isolated HGPIN, whereas the increased PSCA mRNA expression after therapy may be a clinically adverse predictor for cancer onset.
Collapse
Affiliation(s)
- Zhao Zhigang
- Department of Urology, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, China.
| | | |
Collapse
|
34
|
Zhigang Z, Wenlu S. The association of prostate stem cell antigen (PSCA) mRNA expression and subsequent prostate cancer risk in men with benign prostatic hyperplasia following transurethral resection of the prostate. Prostate 2008; 68:190-9. [PMID: 18076024 DOI: 10.1002/pros.20701] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Prior data showed prostate stem cell antigen (PSCA) mRNA expression in benign prostatic hyperplasia (BPH) tissues. The purpose of the present investigation was to determine whether PSCA mRNA expression in resected BPH samples was associated with the subsequent presence of cancer following transurethral resection of the prostate (TURP). METHODS PSCA in situ hybridization was performed on the TURP-resected tissues from 288 patients, who were histopathologically confirmed BPH without cancer. All these patients were continuously followed for 9-70 months postoperatively. Univariate and multivariate cox regression analyses were used to evaluate the predictive performance of PSCA mRNA for subsequent cancer onset following TURP. RESULTS PSCA mRNA was detected in 93/288 (32.3%) of the resected BPH specimens, with a mean positive-labeling cells of 23.8%, in which 22 patients (23.7%) were identified as having PCa on follow-up. Of 195 patients with negative expression for PSCA mRNA 2 (1.0%) were subsequently found with PCa. PSCA mRNA expression levels were directly proportional to higher Gleason score and clinical T stage. Univariate and multivariate cox regression analyses demonstrated that only PSCA mRNA expression was predictive of the subsequent cancer development after TURP, however, PSA velocity was an univariately significant but not multivariately significant predictor. CONCLUSIONS This prospective study identifies PSCA mRNA in BPH as a significant predictor of cancer development after TURP, suggesting that PSCA may be used to identify patients who are at high risk for subsequent cancer onset following TURP for BPH and the PSCA test may be useful when applied for repeat biopsies.
Collapse
Affiliation(s)
- Zhao Zhigang
- Department of Urology, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, China.
| | | |
Collapse
|
35
|
Zhigang Z, Wenlu S. Prostate stem cell antigen (PSCA) mRNA expression in prostatic intraepithelial neoplasia: implications for the development of prostate cancer. Prostate 2007; 67:1143-51. [PMID: 17503471 DOI: 10.1002/pros.20610] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Prior data clearly demonstrated the expression of prostate stem cell antigen (PSCA) mRNA in prostatic intraepithelial neoplasia (PIN) tissues. The purpose of the present investigation was to determine whether PSCA mRNA expression was associated with the presence of cancer in this disease. METHODS One hundred seventeen men were diagnosed with isolated PIN on initial prostate biopsy, 51 with low-grade form (LGPIN), and 66 with high-grade form (HGPIN). PSCA mRNA expression in initial PIN and subsequent cancer was examined by in situ hybridization (ISH). The differences of the PSCA mRNA expression level between the groups were analyzed by the Chi-square and Student's t-test. Univariate and multivariate logistic regression analyses were performed to evaluate the predictive performance of PSCA mRNA. RESULTS PSCA mRNA expression level in 34 subsequent cancers was statistically increased compared with their paired PIN (P < 0.001), with a Gleason's dependence. HGPIN showed statistically high PSCA mRNA expression compared with LGPIN (P < 0.01). PSCA mRNA expression levels were significantly stronger in the initial isolated LGPIN and isolated HGPIN with subsequent cancer than those without (P < 0.001 and P < 0.001, respectively). Multivariate logistic regression analysis demonstrated that only PSCA mRNA was predictive of the onset of subsequent cancer in patients with isolated LGPIN and in those with isolated HGPIN, respectively. CONCLUSIONS Our data identify PSCA mRNA in initial PIN as a significant predictor of subsequent cancer, suggesting that PSCA implies in prostatic tumorigenesis and may be used to identify the patients with isolated PIN who are at high risk for cancer onset in the disease process.
Collapse
Affiliation(s)
- Zhao Zhigang
- Department of Urology, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, China.
| | | |
Collapse
|
36
|
Zhigang Z, Wenlu S. External beam radiotherapy (EBRT) suppressed prostate stem cell antigen (PSCA) mRNA expression in clinically localized prostate cancer. Prostate 2007; 67:653-60. [PMID: 17342746 DOI: 10.1002/pros.20536] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Prostate stem cell antigen (PSCA), a recently identified glycosylphosphatidylinositol (GPI)-anchored cell surface protein belonging to the Thy-1/Ly-6 family of cell surface antigens, is overexpressed in human prostate cancer (PCa). Our recent data indicated that complete androgen ablation could significantly suppress PSCA mRNA expression in primarily organ-confined PCa. The effect of external beam radiotherapy (EBRT), one of the curative treatment options for localized PCa, on tumor PSCA mRNA expression has not been elucidated. The purpose of the present study was to investigate the variations in the expression levels of PSCA mRNA before and after EBRT, and further evaluate the prognostic value of PSCA in this disease. MATERIALS AND METHODS Between January 1999 and June 2005, 87 men with clinically localized adenocarcinoma of the prostate received only EBRT with a total dose of 65-70 Gy for 6.5-7 weeks. PSCA in situ hybridization (ISH) was performed on the cancerous pretreatment biopsy or transurethral resection of prostate (TURP) tissue and post-treatment biopsy tissue of all 87 men, respectively. Tumor cytoplasmic staining of PSCA mRNA was evaluated by two independent pathologists and the differences of PSCA mRNA expression levels between the samples before and after EBRT were analyzed using the Student's t-test. Twenty-four to seventy months continuous follow-up studies after treatment were performed and aimed at assessing the correlation of PSCA mRNA expression level with biochemical relapse and/or distant metastases from the cancer. RESULTS The percent of cells positive for PSCA mRNA by ISH labeling declined from 71.2% (0-93%) +/- 9.7% before EBRT to 30.7% (0-90%) +/- 5.3% after EBRT (P<0.001). Before EBRT, 81 of 87 cases (93.1%) were positive for PSCA mRNA labeling, however, after EBRT the percentage of positive reactivity of PSCA mRNA was decreased to 62 of 81 cases (76.5%), in which 59 men (95.2%) were found without biochemical relapse or distant metastases on follow-up. This decline in PSCA mRNA labeling was directly proportional to higher pretreatment serum PSA level, higher tumor grade (Gleason score), and higher clinical T stage. The rest 19 cases had the increased percentage of cells positive for PSCA mRNA after EBRT, in which 15 cases developed biochemical relapse and/or distant metastases from tumor on follow-up. CONCLUSIONS We found that EBRT for PCa can significantly suppress PSCA mRNA expression and the elevated PSCA mRNA level after EBRT may be a clinically adverse predictor for tumor progression.
Collapse
Affiliation(s)
- Zhao Zhigang
- Department of Urology, The Second Affiliated Hospital, Shantou University Medical College, Guangdong Province, China.
| | | |
Collapse
|
37
|
Abstract
Although prostate-specific antigen (PSA) has evolved as a very useful tool for detection of prostate cancer, there remains an urgent need for more accurate biomarkers to diagnose prostate cancer and predict cancer-related outcomes. Recent advances in the study of proteomics and high throughput techniques have led to the discovery of many potential biomarkers for prostate cancer. This article briefly reviews the current status of PSA testing and discusses several candidate protein biomarkers for prostate cancer, as well as highlighting some recent proteomic discoveries with the potential to supplement or even replace PSA for the diagnosis and prognosis of prostate cancer.
Collapse
Affiliation(s)
- Timothy J Bradford
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
38
|
Schmidt U, Fuessel S, Koch R, Baretton GB, Lohse A, Tomasetti S, Unversucht S, Froehner M, Wirth MP, Meye A. Quantitative multi-gene expression profiling of primary prostate cancer. Prostate 2006; 66:1521-34. [PMID: 16921506 DOI: 10.1002/pros.20490] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND This study describes the evaluation of the expression patterns of prostate-related transcripts in 106 matched prostate tissues from prostatectomies as predictors for prostate cancer (PCa). METHODS Quantitative PCR (QPCR) assays with site-specific hybridization probes were established for four housekeeping genes (GAPDH, HPRT, PBGD, TBP) and nine prostate-related genes (AibZIP, D-GPCR, EZH2, PCA3, PDEF, prostein, PSA, PSCA, TRPM8). RESULTS The relative mRNA expression levels of AibZIP, D-GPCR, EZH2, PCA3, PDEF, PSA, TRPM8 (all P < 0.001) and prostein (P = 0.019) normalized to the TBP reference gene were significantly higher in malignant compared to non-malignant prostate tissues. Employing receiver-operating characteristic (ROC) analyses, PCA3 was the best single tumor marker with the highest area-under-the-curve (AUC = 0.85). A multivariate logit model for the predictability of the tumor was developed, which employed the relative expression levels of EZH2, PCA3, prostein, and TRPM8 and yielded an AUC of 0.90. CONCLUSIONS The transcript marker PCA3 is a powerful predictor of primary PCa but the inclusion of EZH2, prostein, and TRPM8 adds even more to the diagnostic power. The finding of a significantly higher mRNA expression of three different genes (prostein, PSA, TRPM8) in organ-confined tumors compared to non-organ-confined tumors as well as the multi-marker PCa prediction model developed in the retrospective model system on prostatectomies could be of clinical importance for diagnostic purposes, and should be verified in diagnostic biopsies.
Collapse
Affiliation(s)
- Uta Schmidt
- Department of Urology, Technical University of Dresden, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Elsamman EM, Fukumori T, Tanimoto S, Nakanishi R, Takahashi M, Toida K, Kanayama HO. The expression of prostate stem cell antigen in human clear cell renal cell carcinoma: a quantitative reverse transcriptase-polymerase chain reaction analysis. BJU Int 2006; 98:668-73. [PMID: 16925770 DOI: 10.1111/j.1464-410x.2006.06350.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To analyse the gene expression level of prostate stem cell antigen (PSCA) in human clear cell renal cell carcinoma (CC-RCC) and its relationship with conventional clinicopathological manifestations, to evaluate its prognostic value for patient outcome, and to determine the effect of PSCA on the progression of CC-RCC. PATIENTS AND METHODS We quantified PSCA mRNA level in human RCC cell lines (ACHN, A704, KPK-1, Caki-1, and Caki-2) and in 154 surgical tissue samples (81 from CC-RCC, 73 from normal kidney) using real-time reverse transcriptase-polymerase chain reaction (RT-PCR). The findings were analysed in relation to clinicopathological factors. Immunohistochemical expression was examined using confocal laser scanning light-microscopy. RESULTS PSCA was overexpressed in all RCC cell lines. PSCA mRNA levels were significantly higher in CC-RCC than in normal kidney tissue samples (P < 0.001), in G2-G3 than in G1 tumours (P = 0.028), and in advanced disease (T3-T4) than in organ-confined (T1-T2) tumours (P = 0.016). There was significantly higher PSCA mRNA expression in patients with M1 than in those with M0 disease (P = 0.029). Patients in whom the lesions had high PSCA expression levels had a significantly worse prognosis than those with low PSCA expression levels (P = 0.044). Using immunohistochemical analysis there was markedly greater PSCA expression in CC-RCC than in normal kidney, and in advanced-disease high-grade tumours than in organ-confined low-grade tumours. CONCLUSIONS A significant correlation was detected in the gene expression level of PSCA with histological grade, clinicopathological stage and prognosis in CC-RCC. Our data indicate that PSCA is associated with carcinogenesis and progression of CC-RCC.
Collapse
Affiliation(s)
- Essam M Elsamman
- Department of Urology and Department of Anatomy and Cell Biology, The University of Tokushima Graduate School Institute of Health Biosciences, Tokushima, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Bodey B, Bodey B, Siegel SE. Mechanisms and markers of carcinogenesis and neoplastic progression. Expert Opin Biol Ther 2006; 5:1317-32. [PMID: 16197337 DOI: 10.1517/14712598.5.10.1317] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neoplastic transformation evolves over a period of time involving the progression of the cellular immunophenotype (IP) from normal to hyperplastic to dysplastic, and finally, to fully malignant IPs. Superimposed on these changes is the interaction of the initiated cell with its microenvironment, whereby the neoplastically transformed cells, through the regulation or dysregulation of cytoskeletal, integrin, protease and adhesion molecules, develop a novel manner of relation with their surrounding microenvironment. Studies of the neuroendocrine-immune network revealed that the hormonal and cytokine milieu plays an important role impacting the growth and dedifferentiation capabilities of neoplastic cells. This is further affected by the tumour cells themselves determining the constitution of this hormonal microenvironment, allowing the most aggressive and invasive of neoplastically transformed cell clones to promote their own growth and dissemination. The elucidation of the steps of the progression of cancer from premalignant to metastatic and invasive forms is of utmost importance in the differential diagnosis of neoplasms and in the establishment of more efficacious therapeutic regimens. These regimens will certainly begin to take on a more individualised form. The functional characterisation of various human malignancies as to the neoplastically transformed cells' IP, the bases of their interaction with tissue stromal elements, and the molecules involved in the humoral microenvironment of the particular stage of tumour will certainly allow for the better diagnosis, staging, prognostication and treatment of cancers in the future. This paper reviews carcinogenesis from nutritional, genetic and molecular, and humoral aspects, and discusses the importance of tumour markers in the diagnosis and therapeutic management of human cancer.
Collapse
Affiliation(s)
- Bela Bodey
- Department of Pathology, University of Southern California, Keck School of Medicine, Reseda, Los Angeles, CA 91335, USA.
| | | | | |
Collapse
|
41
|
Wang XD, Leow CC, Zha J, Tang Z, Modrusan Z, Radtke F, Aguet M, de Sauvage FJ, Gao WQ. Notch signaling is required for normal prostatic epithelial cell proliferation and differentiation. Dev Biol 2006; 290:66-80. [PMID: 16360140 DOI: 10.1016/j.ydbio.2005.11.009] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Revised: 11/05/2005] [Accepted: 11/07/2005] [Indexed: 12/22/2022]
Abstract
Notch pathway is crucial for stem/progenitor cell maintenance, growth and differentiation in a variety of tissues. Using a transgenic cell ablation approach, we found in our previous study that cells expressing Notch1 are crucial for prostate early development and re-growth. Here, we further define the role of Notch signaling in regulating prostatic epithelial cell growth and differentiation using biochemical and genetic approaches in ex vivo or in vivo systems. Treatment of developing prostate grown in culture with inhibitors of gamma-secretase/presenilin, which is required for Notch cleavage and activation, caused a robust increase in proliferation of epithelial cells co-expressing cytokeratin 8 and 14, lack of luminal/basal layer segregation and dramatically reduced branching morphogenesis. Using conditional Notch1 gene deletion mouse models, we found that inactivation of Notch1 signaling resulted in profound prostatic alterations, including increased tufting, bridging and enhanced epithelial proliferation. Cells within these lesions co-expressed both luminal and basal cell markers, a feature of prostatic epithelial cells in predifferentiation developmental stages. Microarray analysis revealed that the gene expression in a number of genetic networks was altered following Notch1 gene deletion in prostate. Furthermore, expression of Notch1 and its effector Hey-1 gene in human prostate adenocarcinomas were found significantly down-regulated compared to normal control tissues. Taken together, these data suggest that Notch signaling is critical for normal cell proliferation and differentiation in the prostate, and deregulation of this pathway may facilitate prostatic tumorigenesis.
Collapse
Affiliation(s)
- Xi-De Wang
- Department of Molecular Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
As prostate cancer is not a single disease, it is important to identify the pivotal pathway in the patient being treated. The molecular environment is the site of current oncological research to define new therapeutic targets for hormone-refractory disease, offering the potential to eventually individualize treatment through stratification of pathways. Targets may be validated either phenotypically (e.g. androgen receptor, cadherin) or functionally (e.g. prostate cancer-specific genes). In addition, several other candidates are potentially suitable, while others await discovery. Important initial steps have been made in the search for prostate cancer stem cells; identifying stem cells and the stromal, hormonal, and other signalling molecules that influence their behaviour would have important implications for managing prostate cancer. Although individual therapeutic pathways might be ineffective in a particular molecular environment, combinations of approaches might be capable of producing synergistic effects. A multimodal approach thus might be the best solution. Determining where best to search for a molecular target, and validating whether the target is associated with a sufficiently aggressive malignant process to justify further study is difficult, but the potential benefits are enormous.
Collapse
Affiliation(s)
- Jack A Schalken
- Department of Experimental Urology, Radboud University Nijmegen Medical Center, Geert Grooteplein 30, Nijmegen, the Netherlands.
| |
Collapse
|
43
|
|
44
|
Zhigang Z, Wenlu S. Complete androgen ablation suppresses prostate stem cell antigen (PSCA) mRNA expression in human prostate carcinoma. Prostate 2005; 65:299-305. [PMID: 16015594 DOI: 10.1002/pros.20290] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Prostate stem cell antigen (PSCA) is a recently identified glycosylphosphatidylinositol (GPI)-anchored cell surface protein belonging to the Thy-1/Ly-6 family of cell surface antigens. Prior data in prostate cancers indicated that PSCA is directly regulated by androgens and PSCA expression increases with high-tumor grade, advanced stage, extracapsular invasion, and androgen-independent progression. The effect of complete androgen ablation (CAA) on tumor PSCA mRNA expression has not been elucidated. The purpose of the present study was to investigate the variations in the expression levels of PSCA mRNA before and after CAA, and further evaluate the clinically prognostic value of PSCA in human prostate carcinoma. MATERIALS AND METHODS PSCA in situ hybridization (ISH) was performed on the cancerous pretreatment biopsy or transurethral resection of prostate (TURP) tissue of 42 men with primarily organ-confined prostate cancer before CAA, and on their tumor tissue from radical retropubic prostatectomy after CAA with bicalutamide and goserelin acetate for 3 months prior to undergoing radical prostatectomy. Tumor cytoplasmic staining of PSCA mRNA was evaluated by two independent pathologists and the differences of PSCA mRNA expression levels between the samples before and after CAA were analyzed using the Student's t-test. Thirty-six to forty months follow-up studies after radical retropubic prostatectomy were performed and aimed at assessing the correlation of PSCA mRNA expression level with local recurrences or metastases from the cancer. RESULTS The percent of cells positive for PSCA mRNA by ISH labeling declined from 67.3% (0-89%)+/-9.4% before CAA to 33.8% (0-92%)+/-7.7% after CAA (P<0.001). Before CAA, 40 of 42 cases (95.2%) were positive for PSCA mRNA labeling, however, after CAA the percentage of positive reactivity of PSCA mRNA was decreased to 27 of 40 cases (67.5%), in which none was found with local recurrences or distant metastases after radical prostatectomy on follow-up. This decline in PSCA mRNA labeling was dependent on the original tumor grade with Gleason score of <or=6: 19.3%+/-4.7%, Gleason score of 7: 38.8%+/-7.2%, and a Gleason score of >or=8: 73.4%+/-13.8% (P<0.05, respectively). The rest 13 cases had the increased percentage of cells positive for PSCA mRNA after CAA, in which 3 cases were found with local recurrences and 4 cases with distant metastases from tumor on follow-up. CONCLUSIONS Our data demonstrate that CAA for prostate cancer can suppress PSCA mRNA expression with a tumor grade dependence and the increased expression of PSCA mRNA after CAA may be a clinically adverse predictor for tumor recurrences or distant metastases.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Androgen Antagonists/therapeutic use
- Anilides/therapeutic use
- Antigens, Neoplasm
- Biopsy
- Follow-Up Studies
- GPI-Linked Proteins
- Humans
- In Situ Hybridization
- Male
- Membrane Glycoproteins/antagonists & inhibitors
- Membrane Glycoproteins/biosynthesis
- Middle Aged
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/biosynthesis
- Neoplasms, Hormone-Dependent/drug therapy
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Nitriles
- Predictive Value of Tests
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Neoplasm/genetics
- Tosyl Compounds
Collapse
Affiliation(s)
- Zhao Zhigang
- Department of Urology, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, China.
| | | |
Collapse
|
45
|
Gu Z, Yamashiro J, Kono E, Reiter RE. Anti–Prostate Stem Cell Antigen Monoclonal Antibody 1G8 Induces Cell Death In vitro and Inhibits Tumor Growth In vivo via a Fc-Independent Mechanism. Cancer Res 2005; 65:9495-500. [PMID: 16230414 DOI: 10.1158/0008-5472.can-05-2086] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostate stem cell antigen (PSCA), a 123-amino acid cell surface glycoprotein, is highly expressed in both local and metastatic prostate cancers as well as in a large proportion of bladder and pancreatic cancers. PSCA overexpression correlates with a high risk of recurrence after primary therapy for prostate cancer. We have reported previously that anti-PSCA monoclonal antibody (mAb) 1G8 inhibits tumor growth, prevents metastasis, and prolongs the survival of mice inoculated with human prostate cancer cell lines and xenografts. The current study was undertaken to elucidate the mechanism of action of anti-PSCA antibody therapy. In particular, we asked whether antitumor activity resulted from recruitment of an immune response or a direct effect on the tumor cell itself. In vitro assays show that both intact 1G8 and F(ab')2 fragments of 1G8 induce prostate cancer cell death. The anti-PSCA antibody-induced cell death is caspase independent and requires antigen cross-linking. These results were confirmed in in vivo models in which both 1G8 and F(ab')2 fragments were able to inhibit prostate tumor formation and growth equally. These results suggest that the anti-PSCA mAb 1G8 acts by a direct, Fc-independent mechanism to inhibit prostate tumor growth both in vitro and in vivo.
Collapse
Affiliation(s)
- Zhennan Gu
- Department of Urology and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095-1738, USA
| | | | | | | |
Collapse
|