1
|
Vecchiotti D, Clementi L, Cornacchia E, Di Vito Nolfi M, Verzella D, Capece D, Zazzeroni F, Angelucci A. Evidence of the Link between Stroma Remodeling and Prostate Cancer Prognosis. Cancers (Basel) 2024; 16:3215. [PMID: 39335188 PMCID: PMC11430343 DOI: 10.3390/cancers16183215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Prostate cancer (PCa), the most commonly diagnosed cancer in men worldwide, is particularly challenging for oncologists when a precise prognosis needs to be established. Indeed, the entire clinical management in PCa has important drawbacks, generating an intense debate concerning the possibility to individuate molecular biomarkers able to avoid overtreatment in patients with pathological indolent cancers. To date, the paradigmatic change in the view of cancer pathogenesis prompts to look for prognostic biomarkers not only in cancer epithelial cells but also in the tumor microenvironment. PCa ecology has been defined with increasing details in the last few years, and a number of promising key markers associated with the reactive stroma are now available. Here, we provide an updated description of the most biologically significant and cited prognosis-oriented microenvironment biomarkers derived from the main reactive processes during PCa pathogenesis: tissue adaptations, inflammatory response and metabolic reprogramming. Proposed biomarkers include factors involved in stromal cell differentiation, cancer-normal cell crosstalk, angiogenesis, extracellular matrix remodeling and energy metabolism.
Collapse
Affiliation(s)
- Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Letizia Clementi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Emanuele Cornacchia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Mauro Di Vito Nolfi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Adriano Angelucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
2
|
Ben-Eltriki M, Gayle EJ, Walker N, Deb S. Pharmacological Significance of Heme Oxygenase 1 in Prostate Cancer. Curr Issues Mol Biol 2023; 45:4301-4316. [PMID: 37232742 DOI: 10.3390/cimb45050273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Heme oxygenase 1 (HO-1) is a detoxifying antioxidant microsomal enzyme that regulates inflammation, apoptosis, cell proliferation, and angiogenesis in prostate cancer (PCa). This makes HO-1 a promising target for therapeutic prevention and treatment due to its anti-inflammatory properties and ability to control redox homeostasis. Clinical evidence highlights the possible correlation between HO-1 expression and PCa growth, aggressiveness, metastasized tumors, resistance to therapy, and poor clinical outcomes. Interestingly, studies have reported anticancer benefits mediated by both HO-1 induction and inhibition in PCa models. Contrasting evidence exists on the role of HO-1 in PCa progression and possible treatment targets. Herein, we provide an overview of available evidence on the clinical significance of HO-1 signaling in PCa. It appears that the beneficial effects of HO-1 induction or inhibition are dependent on whether it is a normal versus malignant cell as well as the intensity (major vs. minor) of the increase in HO-1 enzymatic activity. The current literature evidence indicates that HO-1 has dual effects in PCa. The amount of cellular iron and reactive oxygen species (ROS) can determine the role of HO-1 in PCa. A major increase in ROS enforces HO-1 to a protective role. HO-1 overexpression may provide cryoprotection to normal cells against oxidative stress via suppressing the expression of proinflammatory genes, and thus offer therapeutic prevention. In contrast, a moderate increase in ROS can lead to the perpetrator role of HO-1, which is associated with PCa progression and metastasis. HO-1 inhibition by xenobiotics in DNA-damaged cells tilts the balance to promote apoptosis and inhibit PCa proliferation and metastasis. Overall, the totality of the evidence revealed that HO-1 may play a dual role in the therapeutic prevention and treatment of PCa.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- Department of Pharmacology and Therapeutics, Clinical Pharmacology Lab, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
- Cochrane Hypertension Review Group, Therapeutic Initiative, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Erysa J Gayle
- College of Biomedical Sciences, Larkin University, 18301 N. Miami Avenue, Miami, FL 33169, USA
| | - Noah Walker
- College of Biomedical Sciences, Larkin University, 18301 N. Miami Avenue, Miami, FL 33169, USA
| | - Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA
| |
Collapse
|
3
|
Zhang W, Lee AM, Jena S, Huang Y, Ho Y, Tietz KT, Miller CR, Su MC, Mentzer J, Ling AL, Li Y, Dehm SM, Huang RS. Computational drug discovery for castration-resistant prostate cancers through in vitro drug response modeling. Proc Natl Acad Sci U S A 2023; 120:e2218522120. [PMID: 37068243 PMCID: PMC10151558 DOI: 10.1073/pnas.2218522120] [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: 10/31/2022] [Accepted: 03/17/2023] [Indexed: 04/19/2023] Open
Abstract
Prostate cancer (PC) is the most frequently diagnosed malignancy and a leading cause of cancer deaths in US men. Many PC cases metastasize and develop resistance to systemic hormonal therapy, a stage known as castration-resistant prostate cancer (CRPC). Therefore, there is an urgent need to develop effective therapeutic strategies for CRPC. Traditional drug discovery pipelines require significant time and capital input, which highlights a need for novel methods to evaluate the repositioning potential of existing drugs. Here, we present a computational framework to predict drug sensitivities of clinical CRPC tumors to various existing compounds and identify treatment options with high potential for clinical impact. We applied this method to a CRPC patient cohort and nominated drugs to combat resistance to hormonal therapies including abiraterone and enzalutamide. The utility of this method was demonstrated by nomination of multiple drugs that are currently undergoing clinical trials for CRPC. Additionally, this method identified the tetracycline derivative COL-3, for which we validated higher efficacy in an isogenic cell line model of enzalutamide-resistant vs. enzalutamide-sensitive CRPC. In enzalutamide-resistant CRPC cells, COL-3 displayed higher activity for inhibiting cell growth and migration, and for inducing G1-phase cell cycle arrest and apoptosis. Collectively, these findings demonstrate the utility of a computational framework for independent validation of drugs being tested in CRPC clinical trials, and for nominating drugs with enhanced biological activity in models of enzalutamide-resistant CRPC. The efficiency of this method relative to traditional drug development approaches indicates a high potential for accelerating drug development for CRPC.
Collapse
Affiliation(s)
- Weijie Zhang
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN55455
- The Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN55455
| | - Adam M. Lee
- The Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN55455
| | - Sampreeti Jena
- The Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN55455
| | - Yingbo Huang
- The Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN55455
| | - Yeung Ho
- Department of Laboratory Medicine and Pathology, The University of Minnesota Medical School, Minneapolis, MN55455
| | - Kiel T. Tietz
- Department of Laboratory Medicine and Pathology, The University of Minnesota Medical School, Minneapolis, MN55455
| | - Conor R. Miller
- Department of Laboratory Medicine and Pathology, The University of Minnesota Medical School, Minneapolis, MN55455
| | - Mei-Chi Su
- The Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN55455
| | - Joshua Mentzer
- The Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN55455
| | - Alexander L. Ling
- The Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN55455
| | - Yingming Li
- Department of Laboratory Medicine and Pathology, The University of Minnesota Medical School, Minneapolis, MN55455
| | - Scott M. Dehm
- Department of Laboratory Medicine and Pathology, The University of Minnesota Medical School, Minneapolis, MN55455
| | - R. Stephanie Huang
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN55455
- The Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN55455
| |
Collapse
|
4
|
Singer D, Ressel V, Stope MB, Bekeschus S. Heat Shock Protein 27 Affects Myeloid Cell Activation and Interaction with Prostate Cancer Cells. Biomedicines 2022; 10:biomedicines10092192. [PMID: 36140293 PMCID: PMC9496253 DOI: 10.3390/biomedicines10092192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/31/2022] [Accepted: 09/04/2022] [Indexed: 11/16/2022] Open
Abstract
Heat shock proteins are cytoprotective molecules induced by environmental stresses. The small heat shock protein 27 (Hsp27) is highly expressed under oxidative stress conditions, mediating anti-oxidative effects and blocking apoptosis. Since medical gas plasma treatment subjects cancer cells to a multitude of reactive oxygen species (ROS), inducing apoptosis and immunomodulation, probable effects of Hsp27 should be investigated. To this end, we quantified the extracellular Hsp27 in two prostate cancer cell lines (LNCaP, PC-3) after gas plasma-induced oxidative stress, showing a significantly enhanced release. To investigate immunomodulatory effects, two myeloid cell lines (THP-1 and HL-60) were also exposed to Hsp27. Only negligible effects on viability, intracellular oxidative milieu, and secretion profiles of the myeloid cells were found when cultured alone. Interestingly, prostate cancer-myeloid cell co-cultures showed altered secretion profiles with a significant decrease in vascular endothelial growth factor (VEGF) release. Furthermore, the myeloid surface marker profiles were changed, indicating an enhanced differentiation in co-culture upon Hsp27 treatment. Finally, we investigated morphological changes, proliferation, and interaction with prostate cancer cells, and found significant alterations in the myeloid cells, supporting the tendency to differentiate. Collectively, our results suggest an ambiguous effect of Hsp27 on myeloid cells in the presence of prostate cancer cells which needs to be further investigated.
Collapse
Affiliation(s)
- Debora Singer
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Verena Ressel
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
- Clinic and Policlinic for Urology, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Matthias B. Stope
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, 53127 Bonn, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
- Correspondence:
| |
Collapse
|
5
|
VEGF Immunoexpression in Prostate Adenocarcinoma. CURRENT HEALTH SCIENCES JOURNAL 2021; 47:89-95. [PMID: 34211753 PMCID: PMC8200605 DOI: 10.12865/chsj.47.01.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/21/2021] [Indexed: 11/18/2022]
Abstract
Angiogenesis is a basic biomolecular mechanism for tumor progression, the vascular endothelial growth factor (VEGF) being one of the main enhancers of this complex process. In this study, we analyzed VEGF-A immunoexpression in 61 prostate adenocarcinomas (PAs), related to the prognostic parameters of the lesions. VEGF scores were higher in PAs that associated serum PSA values above 20ng/ml, in tumors with pure complex or mixed growth patterns, as well as in high-grade and advanced lesions. The results obtained indicate the involvement of VEGF in prostate angiogenesis and the usefulness of the maker for the identification of aggressive lesions.
Collapse
|
6
|
Signaling Pathways That Control Apoptosis in Prostate Cancer. Cancers (Basel) 2021; 13:cancers13050937. [PMID: 33668112 PMCID: PMC7956765 DOI: 10.3390/cancers13050937] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 02/18/2021] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer is the second most common malignancy and the fifth leading cancer-caused death in men worldwide. Therapies that target the androgen receptor axis induce apoptosis in normal prostates and provide temporary relief for advanced disease, yet prostate cancer that acquired androgen independence (so called castration-resistant prostate cancer, CRPC) invariably progresses to lethal disease. There is accumulating evidence that androgen receptor signaling do not regulate apoptosis and proliferation in prostate epithelial cells in a cell-autonomous fashion. Instead, androgen receptor activation in stroma compartments induces expression of unknown paracrine factors that maintain homeostasis of the prostate epithelium. This paradigm calls for new studies to identify paracrine factors and signaling pathways that control the survival of normal epithelial cells and to determine which apoptosis regulatory molecules are targeted by these pathways. This review summarizes the recent progress in understanding the mechanism of apoptosis induced by androgen ablation in prostate epithelial cells with emphasis on the roles of BCL-2 family proteins and "druggable" signaling pathways that control these proteins. A summary of the clinical trials of inhibitors of anti-apoptotic signaling pathways is also provided. Evidently, better knowledge of the apoptosis regulation in prostate epithelial cells is needed to understand mechanisms of androgen-independence and implement life-extending therapies for CRPC.
Collapse
|
7
|
Angiogenesis Inhibition in Prostate Cancer: An Update. Cancers (Basel) 2020; 12:cancers12092382. [PMID: 32842503 PMCID: PMC7564110 DOI: 10.3390/cancers12092382] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/14/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer (PCa), like all other solid tumors, relies on angiogenesis for growth, progression, and the dissemination of tumor cells to other parts of the body. Despite data from in vitro and in vivo preclinical studies, as well as human specimen studies indicating the crucial role played by angiogenesis in PCa, angiogenesis inhibition in clinical settings has not shown significant benefits to patients, thus challenging the inclusion and usefulness of antiangiogenic agents for the treatment of PCa. However, one of the apparent reasons why these antiangiogenic agents failed to meet expectations in PCa can be due to the choice of the antiangiogenic agents, because the majority of these drugs target vascular endothelial growth factor-A (VEGFA) and its receptors. The other relevant causes might be inappropriate drug combinations, the duration of treatment, and the method of endpoint determination. In this review, we will first discuss the role of angiogenesis in PCa growth and progression. We will then summarize the different angiogenic growth factors that influence PCa growth dynamics and review the outcomes of clinical trials conducted with antiangiogenic agents in PCa patients and, finally, critically assess the current status and fate of antiangiogenic therapy in this disease.
Collapse
|
8
|
Oh F, Modiano JF, Bachanova V, Vallera DA. Bispecific Targeting of EGFR and Urokinase Receptor (uPAR) Using Ligand-Targeted Toxins in Solid Tumors. Biomolecules 2020; 10:biom10060956. [PMID: 32630411 PMCID: PMC7356355 DOI: 10.3390/biom10060956] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/31/2022] Open
Abstract
Ligand-targeted toxins (LTTs) are bioengineered molecules which are composed of a targeting component linked to a toxin that induces cell death once the LTT binds its target. Bispecific targeting allows for the simultaneous targeting of two receptors. In this review, we mostly focus on the epidermal growth factor receptor (EGFR) as a target. We discuss the development and testing of a bispecific LTT targeting EGFR and urokinase-type plasminogen activator receptor (uPAR) as two attractive targets implicated in tumor growth and in the regulation of the tumor microvasculature in solid tumors. In vitro and mouse xenograft studies have shown that EGFR-targeted bispecific angiotoxin (eBAT) is effective against human solid tumors. Canine studies have shown that eBAT is both safe and effective against canine hemangiosarcoma, which is physiologically similar to human angiosarcoma. Finding the appropriate dosing strategy and sequencing of eBAT administration, in combination with other therapeutics, are among important factors for future directions. Together, the data indicate that eBAT targets cancer stem cells, it may have a role in inhibiting human tumor vasculature, and its bispecific conformation may have a role in reducing toxicity in comparative oncologic trials in dogs.
Collapse
Affiliation(s)
- Felix Oh
- School of Medicine, Department of Radiation Oncology, University of Minnesota, Minneapolis, MN 55455 USA;
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (J.F.M.); (V.B.)
| | - Jaime F. Modiano
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (J.F.M.); (V.B.)
- College of Veterinary Medicine, Department of Veterinary Clinical Sciences, University of Minnesota, St Paul, MN 55108, USA
| | - Veronika Bachanova
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (J.F.M.); (V.B.)
- School of Medicine, Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel A. Vallera
- School of Medicine, Department of Radiation Oncology, University of Minnesota, Minneapolis, MN 55455 USA;
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (J.F.M.); (V.B.)
- Correspondence: ; Tel.: +61-26-266-664
| |
Collapse
|
9
|
Cytokines and Chemokines as Mediators of Prostate Cancer Metastasis. Int J Mol Sci 2020; 21:ijms21124449. [PMID: 32585812 PMCID: PMC7352203 DOI: 10.3390/ijms21124449] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/16/2022] Open
Abstract
The consequences of prostate cancer metastasis remain severe, with huge impact on the mortality and overall quality of life of affected patients. Despite the convoluted interplay and cross talk between various cell types and secreted factors in the metastatic process, cytokine and chemokines, along with their receptors and signaling axis, constitute important factors that help drive the sequence of events that lead to metastasis of prostate cancer. These proteins are involved in extracellular matrix remodeling, epithelial-mesenchymal-transition, angiogenesis, tumor invasion, premetastatic niche creation, extravasation, re-establishment of tumor cells in secondary organs as well as the remodeling of the metastatic tumor microenvironment. This review presents an overview of the main cytokines/chemokines, including IL-6, CXCL12, TGFβ, CXCL8, VEGF, RANKL, CCL2, CX3CL1, IL-1, IL-7, CXCL1, and CXCL16, that exert modulatory roles in prostate cancer metastasis. We also provide extensive description of their aberrant expression patterns in both advanced disease states and metastatic sites, as well as their functional involvement in the various stages of the prostate cancer metastatic process.
Collapse
|
10
|
Ulgen E, Ozisik O, Sezerman OU. pathfindR: An R Package for Comprehensive Identification of Enriched Pathways in Omics Data Through Active Subnetworks. Front Genet 2019; 10:858. [PMID: 31608109 PMCID: PMC6773876 DOI: 10.3389/fgene.2019.00858] [Citation(s) in RCA: 280] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 08/16/2019] [Indexed: 12/13/2022] Open
Abstract
Pathway analysis is often the first choice for studying the mechanisms underlying a phenotype. However, conventional methods for pathway analysis do not take into account complex protein-protein interaction information, resulting in incomplete conclusions. Previously, numerous approaches that utilize protein-protein interaction information to enhance pathway analysis yielded superior results compared to conventional methods. Hereby, we present pathfindR, another approach exploiting protein-protein interaction information and the first R package for active-subnetwork-oriented pathway enrichment analyses for class comparison omics experiments. Using the list of genes obtained from an omics experiment comparing two groups of samples, pathfindR identifies active subnetworks in a protein-protein interaction network. It then performs pathway enrichment analyses on these identified subnetworks. To further reduce the complexity, it provides functionality for clustering the resulting pathways. Moreover, through a scoring function, the overall activity of each pathway in each sample can be estimated. We illustrate the capabilities of our pathway analysis method on three gene expression datasets and compare our results with those obtained from three popular pathway analysis tools. The results demonstrate that literature-supported disease-related pathways ranked higher in our approach compared to the others. Moreover, pathfindR identified additional pathways relevant to the conditions that were not identified by other tools, including pathways named after the conditions.
Collapse
Affiliation(s)
- Ege Ulgen
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ozan Ozisik
- Department of Computer Engineering, Electrical & Electronics Faculty, Yildiz Technical University, Istanbul, Turkey
| | - Osman Ugur Sezerman
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
11
|
Zhang B, Wu Z, Xie W, Tian D, Chen F, Qin C, Du Z, Tang G, Gao Q, Qiu X, Wu C, Tian J, Hu H. The expression of vasohibin-1 and its prognostic significance in bladder cancer. Exp Ther Med 2017; 14:3477-3484. [PMID: 29042936 PMCID: PMC5639433 DOI: 10.3892/etm.2017.4969] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 05/19/2017] [Indexed: 11/05/2022] Open
Abstract
Angiogenesis is important in the development of solid tumors. Vasohibin-1 (VASH-1) is an endothelium-derived protein that acts as an inhibitor of angiogenesis in many different types of cancer. However, the expression of VASH-1 and its clinical value in bladder cancer remain unknown. The current study analyzed the expression of VASH-1, as well as the expression of the angiogenesis-related factors vascular endothelial growth factor-A, hypoxia inducible factor-1α and cluster of differentiation 34 in bladder cancer tissues from 50 patients using immunohistochemistry. The associations between the expression of these factors and the clinicopathological characteristics of the patients were assessed. The current study demonstrated that VASH-1 is primarily expressed in the cytoplasm of bladder cancer cells and in a fraction of vascular endothelial cells. Furthermore, the expression of VASH-1 was positively associated with the tumor stage (P<0.01), pathological grade (P<0.01) and distant metastasis (P<0.05) but not with patient age or sex (P>0.05). Spearman rank correlation tests indicated that levels of those four factors were positively correlated with each other. Kaplan-Meier analysis indicated that high expression of these four factors was significantly associated with lower 5-year overall survival and progression-free survival rates. Collectively, the results of the current study suggest that VASH-1 is clinically significant in bladder cancer and its high expression may predict the progression and prognosis of patients with bladder cancer. The present study also implies that VASH-1 may be a novel target for vascular targeting therapy.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Department of Ultrasound, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Zhouliang Wu
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Wanqin Xie
- Key Laboratory of Genetics and Birth Health of Hunan, The Family Planning Research Institute of Hunan, Changsha, Hunan 410126, P.R. China
| | - Dawei Tian
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Feiran Chen
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Chuan Qin
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Zhiyong Du
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Gang Tang
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Qiongqiong Gao
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300211, P.R. China
| | - Xiaoyu Qiu
- College of Management and Economics, Tianjin University, Tianjin 300211, P.R. China
| | - Changli Wu
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Jing Tian
- Department of Ultrasound, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Hailong Hu
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| |
Collapse
|
12
|
Huang J, Li Z, Ding Z, Luo Q, Lu S. Different roles of myofibroblasts in the tumorigenesis of nonsmall cell lung cancer. Tumour Biol 2016; 37:15525–15534. [PMID: 26482615 DOI: 10.1007/s13277-015-3862-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 07/28/2015] [Indexed: 11/27/2022] Open
Abstract
Myofibroblasts play a critical role in the cancer cell growth, invasion, and tumor-associated vascularization during the carcinogenesis of nonsmall cell lung cancer (NSCLC), whereas the underlying molecular bases are not completely understood. We isolated Lin-negative, Sca1-low, and CD49e-high myofibroblasts from the NSCLC tissues of the patients and modified the levels of either transforming growth factor β 1 (TGFβ1) or vascular endothelial growth factor A (VEGF-A) in these cells. We found that coculture with TGFβ1-overexpressing myofibroblasts significantly decreased the NSCLC cell growth in an MTT assay through proliferation suppression rather than modulation of cell apoptosis, while significantly increased the NSCLC cell invasiveness in either a transwell migration assay or a scratch wound healing migration assay. However, modulation of TGFβ1 levels in myofibroblasts did not significantly alter vessel formation in a human umbilical vein endothelial cells (HUVECs) transwell collagen gel assay. On the other hand, overexpression of VEGF-A in myofibroblasts significantly increased vessel formation in the HUVECs transwell collagen gel assay. Together, these data suggest that myofibroblasts may regulate cancer cell growth and invasion through TGFβ1 but modulate cancer-associated neovascularization through VEGF-A. Hence, targeting different signaling pathways in myofibroblasts may delicately control NSCLC growth and invasion.
Collapse
Affiliation(s)
- Jia Huang
- Shanghai Lung Tumor Clinical Medical Center, Shanghai Chest Hospital Affiliated To Shanghai Jiaotong University, 241 Huaihai West Road, Shanghai, 200030, China
| | - Ziming Li
- Shanghai Lung Tumor Clinical Medical Center, Shanghai Chest Hospital Affiliated To Shanghai Jiaotong University, 241 Huaihai West Road, Shanghai, 200030, China
| | - Zhengping Ding
- Shanghai Lung Tumor Clinical Medical Center, Shanghai Chest Hospital Affiliated To Shanghai Jiaotong University, 241 Huaihai West Road, Shanghai, 200030, China
| | - Qingquan Luo
- Shanghai Lung Tumor Clinical Medical Center, Shanghai Chest Hospital Affiliated To Shanghai Jiaotong University, 241 Huaihai West Road, Shanghai, 200030, China.
| | - Shun Lu
- Shanghai Lung Tumor Clinical Medical Center, Shanghai Chest Hospital Affiliated To Shanghai Jiaotong University, 241 Huaihai West Road, Shanghai, 200030, China.
| |
Collapse
|
13
|
Köken T, Koca B, Özkurt M, Erkasap N, Kuş G, Karalar M. Apium graveolensExtract Inhibits Cell Proliferation and Expression of Vascular Endothelial Growth Factor and Induces Apoptosis in the Human Prostatic Carcinoma Cell Line LNCaP. J Med Food 2016; 19:1166-1171. [DOI: 10.1089/jmf.2016.0061] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Tülay Köken
- Department of Clinical Biochemistry, School of Medicine, Afyon Kocatepe University, Afyon, Turkey
| | - Buğra Koca
- Department of Clinical Biochemistry, School of Medicine, Afyon Kocatepe University, Afyon, Turkey
| | - Mete Özkurt
- Department of Physiology, School of Medicine, Osmangazi University, Eskişehir, Turkey
| | - Nilüfer Erkasap
- Department of Physiology, School of Medicine, Osmangazi University, Eskişehir, Turkey
| | - Gökhan Kuş
- Department of Physiology, School of Medicine, Osmangazi University, Eskişehir, Turkey
| | - Mustafa Karalar
- Department of Urology, School of Medicine, Afyon Kocatepe University, Afyon, Turkey
| |
Collapse
|
14
|
Posadas EM, Ahmed RS, Karrison T, Szmulewitz RZ, O’Donnell PH, Wade JL, Shen J, Gururajan M, Sievert M, Stadler WM. Saracatinib as a metastasis inhibitor in metastatic castration-resistant prostate cancer: A University of Chicago Phase 2 Consortium and DOD/PCF Prostate Cancer Clinical Trials Consortium Study. Prostate 2016; 76:286-93. [PMID: 26493492 PMCID: PMC4904773 DOI: 10.1002/pros.23119] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 10/14/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND Fyn is a kinase that is upregulated in a subset of metastatic castration-resistant prostate cancer. Saracatinib potently inhibits Fyn activation. We have noted a relationship between Fyn expression and directional motility, a cellular process related to metastasis. As such we hypothesized that treatment with saracatinib would increase the time required to develop new metastatic lesions. METHODS Patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel were eligible for enrollment. This study was executed as a randomized discontinuation trial. During a lead-in phase of two 28-Day cycles, all patients received saracatinib. Afterward, patients with radiographically stable disease were randomized to either saracatinib or placebo. Patients continued treatment until evidence of new metastasis. RESULTS Thirty-one patients were treated. Only 26% of patients had stable disease after 8 weeks and thus proceeded to randomization. This required early termination of the study for futility. The 70% of patients who progressed after the lead-in phase exhibited expansion of existing lesions or decompensation due to clinical progression without new metastatic lesions. Fatigue was reported in more than 25% of patients (all grades) with only two patients experiencing grade 3 toxicity. Other grade 3 adverse events included dehydration, thrombocytopenia, and weakness. CONCLUSIONS This study was unable to determine if saracatinib had potential as metastasis inhibitor. Metastasis inhibition by saracatinib may still be viable in an earlier time in the disease history.
Collapse
Affiliation(s)
- Edwin M. Posadas
- Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Division of Hematology/Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Correspondence to: Edwin M. Posadas, MD, FACP, Medical Director, Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA.
| | - Rafi S. Ahmed
- Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Division of Hematology/Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Theodore Karrison
- Biostatistics, Department of Health Service, University of Chicago, Chicago, Illinois
| | - Russell Z. Szmulewitz
- Genitourinary Oncology Program, Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Peter H. O’Donnell
- Genitourinary Oncology Program, Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - James L. Wade
- Cancer Care Specialists of Central Illinois, S.C, Decatur, Illinois
| | - James Shen
- Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Division of Hematology/Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Murali Gururajan
- Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Margarit Sievert
- Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Walter M. Stadler
- Genitourinary Oncology Program, Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
15
|
Qi WX, Fu S, Zhang Q, Guo XM. Efficacy and toxicity of anti-VEGF agents in patients with castration-resistant prostate cancer: a meta-analysis of prospective clinical studies. Asian Pac J Cancer Prev 2015; 15:8177-82. [PMID: 25339002 DOI: 10.7314/apjcp.2014.15.19.8177] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Blocking angiogenesis by targeting vascular endothelial growth factor (VEGF) signaling pathway to inhibit tumor growth has proven to be successful in treating a variety of different metastatic tumor types, including kidney, colon, ovarian, and lung cancers, but its role in castration-resistant prostate cancer (CRPC) is still unknown. We here aimed to determine the efficacy and toxicities of anti-VEGF agents in patients with CRPC. MATERIALS AND METHODS The databases of PubMed, Web of Science and abstracts presented at the American Society of Clinical Oncology up to March 31, 2014 were searched for relevant articles. Pooled estimates of the objective response rate (ORR) and prostate-specific antigen (PSA) response rate (decline ≥50%) were calculated using the Comprehensive Meta-Analysis (version 2.2.064) software. Median weighted progression- free survival (PFS) and overall survival (OS) time for anti-VEGF monotherapy and anti-VEGF-based doublets were compared by two-sided Student's t test. RESULTS A total of 3,841 patients from 19 prospective studies (4 randomized controlled trials and 15 prospective nonrandomized cohort studies) were included for analysis. The pooled ORR was 12.4% with a higher response rate of 26.4% (95%CI, 13.6-44.9%) for anti-VEGF-based combinations vs. 6.7% (95%CI, 3.5-12.7%) for anti-VEGF alone (p=0.004). Similarly, the pooled PSA response rate was 32.4% with a higher PSA response rate of 52.8% (95%CI: 40.2-65.1%) for anti-VEGF-based combinations vs. 7.3% (95%CI, 3.6-14.2%) for anti-VEGF alone (p<0.001). Median PFS and OS were 6.9 and 22.1 months with weighted median PFS of 5.6 vs. 6.9 months (p<0.001) and weighted median OS of 13.1 vs. 22.1 months (p<0.001) for anti-VEGF monotherapy vs. anti-VEGF-based doublets. CONCLUSIONS With available evidence, this pooled analysis indicates that anti-VEGF monotherapy has a modest effect in patients with CRPC, and clinical benefits gained from anti-VEGF-based doublets appear greater than anti-VEGF monotherapy.
Collapse
Affiliation(s)
- Wei-Xiang Qi
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China E-mail :
| | | | | | | |
Collapse
|
16
|
Cardoso HJ, Vaz CV, Correia S, Figueira MI, Marques R, Maia CJ, Socorro S. Paradoxical and contradictory effects of imatinib in two cell line models of hormone-refractory prostate cancer. Prostate 2015; 75:923-935. [PMID: 25786656 DOI: 10.1002/pros.22976] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 01/13/2015] [Indexed: 01/26/2023]
Abstract
BACKGROUND Imatinib mesylate is a chemotherapeutic drug that inhibits the tyrosine kinase activity of c-KIT and has been successfully used to treat leukemias and some solid tumors. However, its application for treatment of hormone-refractory prostate cancer (HRPC) has shown modest effectiveness and did not follow the outcomes in cultured cells or animal models. Moreover, the molecular pathways by which imatinib induces cytotoxicity in prostate cancer cells are poorly characterized. METHODS Two cell line models of HRPC (DU145 and PC3) were exposed to 20 μM of imatinib for 6-72 hr. MTS assay was used to assess cell viability during the course of experiment. Gene expression analysis of c-KIT, cell-cycle and apoptosis regulators, and angiogenic factors was determined by means of real-time PCR, western blot, and/or immunocytochemistry. The enzymatic activity of the apoptosis effector, caspase-3, was determined by a colorimetric assay. RESULTS Imatinib significantly decreased the viability of DU145 cells but paradoxically augmented the viability of PC3 cells. DU145 cells displayed diminished expression of anti-apoptotic Bcl-2 protein and augmented levels of caspase-8 and -9, as well as, increased enzymatic activity of caspase-3 in response to imatinib. No differences existed on the expression levels of apoptosis-related proteins in PC3 cells treated with imatinib, though the activity of caspase-3 was decreased. The mRNA levels of angiogenic factor VEGF were decreased in DU145-treated cells, whereas an opposite effect was seen in PC3. In addition, it was shown that DU145 and PC3 cells present a differential expression of c-KIT protein variants. CONCLUSION DU145 and PC3 cells displayed a contradictory behavior in response to imatinib, which was underpinned by a distinct expression pattern (or activity) of target regulators of cell-cycle, apoptosis, and angiogenesis. The paradoxical effect of imatinib in PC3 cells may be related with the differential expression of c-KIT protein variants. Moreover, the present findings helped to understand the discrepancies in the efficacy of imatinib as therapeutic option in HRPC.
Collapse
Affiliation(s)
- Henrique J Cardoso
- CICS-UBI, Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | | | | | | | | | | | | |
Collapse
|
17
|
Gao Q, Zheng J, Yao X, Peng B. Adiponectin inhibits VEGF-A in prostate cancer cells. Tumour Biol 2015; 36:4287-92. [PMID: 25586350 DOI: 10.1007/s13277-015-3067-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/05/2015] [Indexed: 12/25/2022] Open
Abstract
A role of adiponectin in tumorigenesis has recently been appreciated. Although plasma adiponectin levels in subjects with prostate cancer have been found to be significantly lower than in subjects with benign prostatic hyperplasia or in normal healthy controls, the underlying molecular mechanisms remain unknown. Here, we not only detected significant decreases in plasma adiponectin levels in prostate cancer patients, but also showed significant decreases in adiponectin receptor I (AdipoR1) levels in the resected prostate cancer specimen. Prostate cancer cell lines examined in the current study had all lower levels of adiponectin and AdipoR1, compared to normal healthy prostate tissue. Moreover, overexpression of adiponectin in prostate cancer cells decreased production of vascular endothelial growth factor A (VEGF-A), while adiponectin depletion increased VEGF-A. Furthermore, adiponectin seemed to activate AMPK/TSC2 to inhibit mTor-mediated activation of VEGF-A. Taken together, our data suggest that adiponectin may play an essential role in suppressing growth of prostate cancer cells through inhibition of VEGF-A-mediated cancer neovascularization.
Collapse
Affiliation(s)
- Qiruo Gao
- Department of Urology, Shanghai 10th People's Hospital, Tongji University School of Medicine, 301 Yanchangzhong Road, Shanghai, 200072, China
| | | | | | | |
Collapse
|
18
|
Lapuk AV, Volik SV, Wang Y, Collins CC. The role of mRNA splicing in prostate cancer. Asian J Androl 2014; 16:515-21. [PMID: 24830689 PMCID: PMC4104073 DOI: 10.4103/1008-682x.127825] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 02/12/2014] [Indexed: 12/23/2022] Open
Abstract
Alternative splicing (AS) is a crucial step in gene expression. It is subject to intricate regulation, and its deregulation in cancer can lead to a wide array of neoplastic phenotypes. A large body of evidence implicates splice isoforms in most if not all hallmarks of cancer, including growth, apoptosis, invasion and metastasis, angiogenesis, and metabolism. AS has important clinical implications since it can be manipulated therapeutically to treat cancer and represents a mechanism of resistance to therapy. In prostate cancer (PCa) AS also plays a prominent role and this review will summarize the current knowledge of alternatively spliced genes with important functional consequences. We will highlight accumulating evidence on AS of the components of the two critical pathways in PCa: androgen receptor (AR) and phosphoinositide 3-kinase (PI3K). These observations together with data on dysregulation of splice factors in PCa suggest that AR and PI3K pathways may be interconnected with previously unappreciated splicing regulatory networks. In addition, we will discuss several lines of evidence implicating splicing regulation in the development of the castration resistance.
Collapse
Affiliation(s)
- Anna V Lapuk
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Stanislav V Volik
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada
| | - Colin C Collins
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
19
|
Cullu N, Kantarci M, Ogul H, Adanur S, Okur A, Koc E, Pirimoglu B, Karaca L, Kizrak Y, Polat O. Feasibility study of CT perfusion imaging for prostate carcinoma. Eur Radiol 2014; 24:2236-44. [PMID: 24863884 DOI: 10.1007/s00330-014-3212-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 03/25/2014] [Accepted: 04/30/2014] [Indexed: 01/01/2023]
Abstract
OBJECTIVE The aim of this feasibility study was to obtain initial data with which to assess the efficiency of perfusion CT imaging (CTpI) and to compare this with magnetic resonance imaging (MRI) in the diagnosis of prostate carcinoma. MATERIALS AND METHODS This prospective study involved 25 patients with prostate carcinoma undergoing MRI and CTpI. All analyses were performed on T2-weighted images (T2WI), apparent diffusion coefficient (ADC) maps, diffusion-weighted images (DWI) and CTp images. We compared the performance of T2WI combined with DWI and CTp alone. The study was approved by the local ethics committee, and written informed consent was obtained from all patients. RESULTS Tumours were present in 87 areas according to the histopathological results. The diagnostic performance of the T2WI+DWI+CTpI combination was significantly better than that of T2WI alone for prostate carcinoma (P < 0.001). The diagnostic value of CTpI was similar to that of T2WI+DWI in combination. There were statistically significant differences in the blood flow and permeability surface values between prostate carcinoma and background prostate on CTp images. CONCLUSION CTp may be a valuable tool for detecting prostate carcinoma and may be preferred in cases where MRI is contraindicated. If this technique is combined with T2WI and DWI, its diagnostic value is enhanced. KEY POINTS Perfusion CT is a helpful technique for prostate carcinoma diagnosis. •Colour maps allow easy and rapid visual assessment of the functional changes. Colour maps of prostate carcinoma provide information about in vivo tumoral vascularity. CTp images may be added into routine radiological examinations. CTp provides guidance for histopathological correlation if biopsy is scheduled.
Collapse
Affiliation(s)
- Nesat Cullu
- Department of Radiology, Muğla Sıtkı Koçman University, School of Medicine, Muğla, Turkey
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Antiangiogenic therapy effects on age-associated matrix metalloproteinase-9 (MMP-9) and insulin-like growth factor receptor-1 (IGFR-1) responses: a comparative study of prostate disorders in aged and TRAMP mice. Histochem Cell Biol 2014; 142:269-84. [PMID: 24562790 DOI: 10.1007/s00418-014-1193-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2014] [Indexed: 10/25/2022]
Abstract
Senescence is associated with hormonal imbalance and prostatic disorders. Angiogenesis is fundamental for the progression of malignant lesions and is a promising target for prostate cancer treatment. The aim was to characterize matrix metalloproteinase-9 (MMP-9) and insulin-like growth factor receptor-1 (IGFR-1) responses in the prostate during senescence and following antiangiogenic and/or androgen ablation therapies, comparing them to cancer progression features in TRAMP mice. Aged male mice (52-week-old FVB) were submitted to antiangiogenic treatments with SU5416 (6 mg/kg; i.p.) and/or TNP-470 (15 mg/kg; s.c). Finasteride (20 mg/kg; s.c.) was administered alone or associated to both inhibitors. Dorsolateral prostate was collected for light microscopy, and immunohistochemistry and Western blotting collected for MMP-9 and IGFR-1. Senescence led to inflammation and different proliferative lesions in the prostate, as well as to increased MMP-9 and IGFR-1, resembling TRAMP mice prostatic microenvironment. Antiangiogenic therapies promoted recovery and/or interruption of age-associated alterations, presenting differential effects on the molecules studied. SU5416 acted mainly on MMP-9, whereas TNP-470 showed its best influence on IGFR-1 levels. Finasteride administration, alone or in combination with antiangiogenic agents, also resulted in regression of inflammation and neoplastic lesions, besides having a negative modulatory effect on both MMP-9 and IGFR-1. We concluded that stimulated tissue remodeling and proliferative processes during senescence predisposed the prostate to malignant disorders. The combination of different agents was more effective to minimize prostatic imbalance during this period, probably due to the differential action of each drug on factors involved in cell proliferation and extracellular matrix remodeling, resulting in a broader spectrum of effects following the combined treatment.
Collapse
|
21
|
Wen J, Zhao Y, Li J, Weng C, Cai J, Yang K, Yuan H, Imperato-McGinley J, Zhu YS. Suppression of DHT-induced paracrine stimulation of endothelial cell growth by estrogens via prostate cancer cells. Prostate 2013; 73:1069-1081. [PMID: 23423946 PMCID: PMC3923318 DOI: 10.1002/pros.22654] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 01/23/2013] [Indexed: 11/08/2022]
Abstract
BACKGROUND Androgen modulation of angiogenesis in prostate cancer may be not directly mediated by androgen receptor (AR) as AR is not detected in the prostatic endothelial cells. METHODS We examined the paracrine stimulation of cell proliferation by prostate tumor cells and its modulation by androgen and estrogens in a murine endothelial cell line (MEC) that does not express AR. RESULTS Tumor cell conditioned media (TCM) collected from LAPC-4 or LNCaP prostatic tumor cells produced a time- and concentration-dependent induction of cell growth in MECs, which was parallel to the VEGF concentration in the TCM. This TCM-induced cell growth in MECs was enhanced by the treatment of prostatic tumor cells with dihydrotestosterone (DHT). Both the TCM-stimulation and DHT-enhancement effects in MECs were completely blocked by SU5416, a specific VEGF receptor antagonist. Co-administration of 17α-estradiol or 17β-estradiol with DHT in prostatic tumor cells completely inhibited the DHT-enhancement effect while treatment with DHT, 17α-estradiol or 17β-estradiol did not produce any significant direct effect in MECs. Moreover, administration of 17α-estradiol or 17β-estradiol in xenograft animals with LAPC-4 or LNCaP prostate tumor significantly decreased the microvessel number in the tumor tissues. CONCLUSIONS Our study indicated that prostate tumor cells regulate endothelial cell growth through a paracrine mechanism, which is mainly mediated by VEGF; and DHT is able to modulate endothelial cell growth via tumor cells, which is inhibited by 17α-estradiol and 17β-estradiol. Thus, both17α-estradiol and 17β-estradiol are potential agents for anti-angiogenesis therapy in androgen-responsive prostate cancer.
Collapse
Affiliation(s)
- Juan Wen
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
- The Center of Clinical Pharmacology, Central South
University, Changsha, China
| | - Yuan Zhao
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
| | - Jinghe Li
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
| | - Chunyan Weng
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
- The Center of Clinical Pharmacology, Central South
University, Changsha, China
| | - Jingjing Cai
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
- The Center of Clinical Pharmacology, Central South
University, Changsha, China
| | - Kan Yang
- Department of Cardiology of the Third Xiangya Hospital,
Central South University, Changsha, China
| | - Hong Yuan
- The Center of Clinical Pharmacology, Central South
University, Changsha, China
| | | | - Yuan-Shan Zhu
- Department of Medicine/Endocrinology, Weill Cornell Medical
College, NewYork, NewYork
- The Institute of Clinical Pharmacology, Central South
University, Changsha, China
| |
Collapse
|
22
|
Lai CS, Li S, Miyauchi Y, Suzawa M, Ho CT, Pan MH. Potent anti-cancer effects of citrus peel flavonoids in human prostate xenograft tumors. Food Funct 2013; 4:944-9. [PMID: 23673480 DOI: 10.1039/c3fo60037h] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Prostate cancer is one of the most prevalent malignancies and is the second leading cause of cancer-related deaths in men. Fruit and vegetable consumption is a novel, non-toxic therapeutic approach that can be used to prevent and treat prostate cancer. Citrus peels and their extracts have been reported to have potent pharmacological activities and health benefits due to the abundance of flavonoids in citrus fruits, particularly in the peels. Our previous studies demonstrated that oral administration of Gold Lotion (GL), an extract of multiple varieties of citrus peels containing abundant flavonoids, including a large percentage of polymethoxyflavones (PMFs), effectively suppressed azoxymethane (AOM)-induced colonic tumorigenesis. However, the efficacy of GL against prostate cancer has not yet been investigated. Here, we explored the anti-tumor effects of GL using a human prostate tumor xenograft mouse model. Our data demonstrated that treatment with GL by both intraperitoneal (i.p.) injection and oral administration dramatically reduced both the weights (57%-100% inhibition) and volumes (78%-94% inhibition) of the tumors without any observed toxicity. These inhibitory effects were accompanied by mechanistic down-regulation of the protein levels of inflammatory enzymes (inducible nitric oxide synthase, iNOS and cyclooxygenase-2, COX-2), metastasis (matrix metallopeptidase-2, MMP-2 and MMP-9), angiogenesis (vascular endothelial growth factor, VEGF), and proliferative molecules, as well as by the induction of apoptosis in prostate tumors. Our findings suggest that GL is an effective anti-cancer agent that may potentially serve as a novel therapeutic option for prostate cancer treatment.
Collapse
Affiliation(s)
- Ching-Shu Lai
- Department of Seafood Science, National Kaohsiung Marine University, No.142, Haijhuan Rd., Nanzih District, Kaohsiung 81143, Taiwan
| | | | | | | | | | | |
Collapse
|
23
|
Perfusion MDCT of Prostate Cancer: Correlation of Perfusion CT Parameters and Immunohistochemical Markers of Angiogenesis. AJR Am J Roentgenol 2012; 199:1042-8. [DOI: 10.2214/ajr.11.8267] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
24
|
The important roles of RET, VEGFR2 and the RAF/MEK/ERK pathway in cancer treatment with sorafenib. Acta Pharmacol Sin 2012; 33:1311-8. [PMID: 22941289 DOI: 10.1038/aps.2012.76] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
AIM To elucidate the roles of receptor tyrosine kinases RET and VEGFR2 and the RAF/MEK/ERK signaling cascade in cancer treatment with sorafenib. METHODS The cell lines A549, HeLa, and HepG2 were tested. The enzyme activity was examined under cell-free conditions using 384-well microplate assays. Cell proliferation was evaluated using the Invitrogen Alarmar Blue assay. Gene expression was analyzed using the Invitrogen SYBR Green expression assays with a sequence detection system. Protein expression analysis was performed using Western blotting. RESULTS Sorafenib potently suppressed the activities of cRAF, VEGFR2, and RET with IC(50) values of 20.9, 4 and 0.4 nmol/L, respectively. Sorafenib inhibited cRAF, VEGFR2, and RET via non-ATP-competitive, ATP-competitive and mixed-type modes, respectively. In contrast, sorafenib exerted only moderate cytotoxic effects on the proliferation of the 3 cell lines. The IC(50) values for inhibition of A549, HeLa, and HepG2 cells were 8572, 4163, and 8338 nmol/L, respectively. In the 3 cell lines, sorafenib suppressed the cell proliferation mainly by blocking the MEK/ERK downstream pathway at the posttranscriptional level, which in turn regulated related gene expression via a feed-back mechanism. CONCLUSION This study provides novel evidence that protein kinases RET and VEGFR2 play crucial roles in cancer treatment with sorafenib.
Collapse
|
25
|
Najy AJ, Jung YS, Won JJ, Conley-LaComb MK, Saliganan A, Kim CJ, Heath E, Cher ML, Bonfil RD, Kim HRC. Cediranib inhibits both the intraosseous growth of PDGF D-positive prostate cancer cells and the associated bone reaction. Prostate 2012; 72:1328-38. [PMID: 22213159 PMCID: PMC3369116 DOI: 10.1002/pros.22481] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 12/04/2011] [Indexed: 12/31/2022]
Abstract
BACKGROUND The major cause of death in prostate cancer (PCa) cases is due to distant metastatic lesions, with the bone being the most prevalent site for secondary colonization. Utilization of small molecule inhibitors to treat bone metastatic PCa have had limited success either as monotherapies or in combination with other chemotherapeutics due to intolerable toxicities. In the current study, we developed a clinically relevant in vivo intraosseous tumor model overexpressing the platelet-derived growth factor D (PDGF D) to test the efficacy of a newly characterized vascular endothelial growth factor receptor (VEGFR)/PDGFR inhibitor, cediranib (also called AZD2171). METHODS An intratibial-injection model was established utilizing DU145 cells with or without increased PDGF D expression. Tumor-bearing mice were treated by daily gavage administration of cediranib and/or weekly i.p. injection of docetaxel for 7 weeks. Tibiae were monitored by in vivo/ex vivo X-rays and histomorphometry analysis was performed to estimate tumor volume and tumor-associated trabecular bone growth. RESULTS Cediranib reduced intraosseous growth of prostate tumors as well as tumor-associated bone responses. When compared to the standard chemotherapeutic agent docetaxel, cediranib exhibited a stronger inhibition of tumor-associated bone response. The efficacy of cediranib was further enhanced when the drug was co-administered with docetaxel. Importantly, the therapeutic benefits of cediranib and docetaxel are more prominent in intraosseous prostate tumors overexpressing PDGF D. CONCLUSION These novel findings support the utilization of cediranib, either alone or in combination with docetaxel, to treat bone metastatic PCa exhibiting PDGF D expression.
Collapse
Affiliation(s)
- Abdo J. Najy
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Young Suk Jung
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Joshua J. Won
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - M. Katie Conley-LaComb
- Department of Urology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Allen Saliganan
- Department of Urology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Chong Jai Kim
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Elisabeth Heath
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Michael L. Cher
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Urology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - R. Daniel Bonfil
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Urology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Hyeong-Reh Choi Kim
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
- To whom correspondence should be addressed: Department of Pathology, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA, Tel: 313-577-2407, Fax: 313-577-0057,
| |
Collapse
|
26
|
Abstract
The therapeutic approach to advanced prostate cancer has seen greater changes in the last 7 years than it did in the preceding 70. Although only one of the newly approved agents that improve overall survival is a targeted agent, it is a validation of the method of pathway analysis and drug design in delivering novel, clinically usable agents. As our knowledge of the molecular circuitry of tumour invasion, metastases and treatment resistance has become more refined, the number of new, potentially useful, targets has grown exponentially. This is reflected in the vast array of diverse targeted agents that are currently being evaluated in human trials. In this review, we briefly describe some of the key pathways that are involved in the evolution of the prostate cancer 'lethal phenotype', and review the clinical activity of some of the newly approved targeted therapies or those in advanced phases of clinical development.
Collapse
Affiliation(s)
- Niall M Corcoran
- Department of Surgery, University of Melbourne, Royal Melbourne Hospital, Parkville, Victoria, Australia.
| | | |
Collapse
|
27
|
Desiniotis A, Kyprianou N. Advances in the design and synthesis of prazosin derivatives over the last ten years. Expert Opin Ther Targets 2011; 15:1405-18. [PMID: 22148952 DOI: 10.1517/14728222.2011.641534] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Mechanistic, translational and pharmacological studies led to the identification and discovery of the preferred localization, binding characteristics, structure and functional properties of α1-adrenoceptor (α1-AR) subtypes in the bladder neck, bladder and prostate gland. The evidence gathered on α1-ARs, provided a molecular platform for the development of subtype-selective antagonists, resulting in more effective approaches targeting those receptors for the treatment of outlet bladder obstruction and benign prostate hyperplasia. AREAS COVERED Advances over the last decade in the design and optimization of Prazosin, Doxazosin and Terazosin quinazoline-based derivatives as α1-AR antagonists. Evidence on the metabolic and growth interference action by these agents, in addition to their smooth-muscle-relaxing effects. The new action recognition emerges from data on the inhibitory effect of quinazoline-based antagonists on primary tumor growth and progression to metastasis. In addition to the cellular findings in the prostate, functional validation and therapeutic effects of selected lead pharmaceutically optimized derivatives in the context of impairing vascularity and triggering tumor apoptosis. EXPERT OPINION Knowledge on targeting intracellular signalling pathways driving the cellular response via an α1-AR-dependent and independent antagonistic action, must be invested towards the optimization of new agents that while bypassing AR, exhibit improved pharmacological efficacy against human cancer.
Collapse
Affiliation(s)
- Andreas Desiniotis
- University of Kentucky Medical Center, University of Kentucky, College of Medicine, Department of Surgery, Combs Cancer Research Building, Room 306, Lexington, KY 40536, USA
| | | |
Collapse
|
28
|
Oral/metronomic cyclophosphamide-based chemotherapy as option for patients with castration-refractory prostate cancer – Review of the literature. Cancer Treat Rev 2011; 37:444-55. [DOI: 10.1016/j.ctrv.2010.12.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 12/08/2010] [Accepted: 12/18/2010] [Indexed: 11/21/2022]
|
29
|
Yu J, Liu F, Sun Z, Sun M, Sun S. The enhancement of radiosensitivity in human esophageal carcinoma cells by thalidomide and its potential mechanism. Cancer Biother Radiopharm 2011; 26:219-27. [PMID: 21539454 DOI: 10.1089/cbr.2010.0897] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
To investigate the effects of thalidomide on the radiosensitivity of human esophageal cancer cells (TE1 cells) and the potential mechanism underlying these effects. The effects of thalidomide on proliferation of TE1 cells were determined by Methyl thiazolyl tetrazolium assay. The multitarget click model was used to delineate the survival curve using a colony-forming assay, and the radiosensitivity was determined after TE1 cells were treated by thalidomide and/or X-ray radiation. The cell cycle was detected using flow cytometry. Our results are as follows: thalidomide alone suppressed the proliferation of TE1 cells in a dose- and time-dependent manner. The suppressive effects were enhanced by prolonged duration or elevated concentration of thalidomide. However, thalidomide did not affect the cell cycle of TE1 cells. The expression of vascular endothelial growth factor (VEGF) mRNA and protein was suppressed after treatment with thalidomide alone in a dose-dependent manner. Synergistic suppressive effects on VEGF expression were observed after administration of thalidomide and X-ray exposure. In conclusion, thalidomide was able to enhance the radiosensitivity of TE1 cells in vitro, which could be closely related to its suppressive effects on the expression of VEGF in TE1 cells, but had no obvious effects on the cell cycle.
Collapse
Affiliation(s)
- Jingping Yu
- College of Radiological Medicine and Public Health, Soochow University, Suzhou, China
| | | | | | | | | |
Collapse
|
30
|
Phase II Study of Pomalidomide in Patients with Castration-Resistant Prostate Cancer. Cancers (Basel) 2011; 3:3449-60. [PMID: 24212963 PMCID: PMC3759205 DOI: 10.3390/cancers3033449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 08/26/2011] [Accepted: 08/29/2011] [Indexed: 12/03/2022] Open
Abstract
Pomalidomide is a distinct immunomodulatory agent that also displays anti-proliferative and proapoptotic activity. The purpose of this study was to assess the efficacy and safety of pomalidomide for the treatment of chemotherapy-naïve patients with metastatic castration-resistant prostate cancer (CRPC). Methods: Pomalidomide was administered orally in doses of 1 or 2 mg/day without interruption. Follow ups were conducted every 4 weeks with evaluation of study outcomes at 12 weeks. The principal study outcomes were PSA response, time to progression (TTP) using RECIST, overall survival (OS), and safety. A total of 32 patients were enrolled: 15 in the 1 mg/day cohort (median baseline PSA level of 12.30 ng/mL [0.8–236.0]), and 17 in the 2 mg/day cohort (median baseline PSA level of 12.50 ng/mL [0.6–191.8]). Results: In the 1 mg cohort disease was stabilized for ≥28 days in eight patients, and median TTP was 2.90 months. In the 2 mg cohort, PSA decreased ≥50% in three patients, disease was stabilized for ≥28 days in seven patients, and median TTP was 5.87 months. Toxicity in both cohorts was predominantly grade 1 or 2; 2 grade 3 toxicity (fatigue) occurred in the 1 mg cohort, and 5 grade 3 toxicities (chest pain, diarrhea, epigastric pain, impaction, pain) occurred in the 2 mg cohort. One grade 4 toxicity of cardiac ischemia occurred. Conclusions: Pomalidomide shows promising activity in patients with CRPC and has an acceptable safety profile.
Collapse
|
31
|
Huo Q, Colon J, Cordero A, Bogdanovic J, Baker CH, Goodison S, Pensky MY. A facile nanoparticle immunoassay for cancer biomarker discovery. J Nanobiotechnology 2011; 9:20. [PMID: 21605409 PMCID: PMC3127990 DOI: 10.1186/1477-3155-9-20] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2011] [Accepted: 05/23/2011] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Gold nanoparticles (AuNPs) scatter light intensely at or near their surface plasmon wavelength region. Using AuNPs coupled with dynamic light scattering (DLS) detection, we developed a facile nanoparticle immunoassay for serum protein biomarker detection and analysis. A serum sample was first mixed with a citrate-protected AuNP solution. Proteins from the serum were adsorbed to the AuNPs to form a protein corona on the nanoparticle surface. An antibody solution was then added to the assay solution to analyze the target proteins of interest that are present in the protein corona. The protein corona formation and the subsequent binding of antibody to the target proteins in the protein corona were detected by DLS. RESULTS Using this simple assay, we discovered multiple molecular aberrations associated with prostate cancer from both mice and human blood serum samples. From the mice serum study, we observed difference in the size of the protein corona and mouse IgG level between different mice groups (i.e., mice with aggressive or less aggressive prostate cancer, and normal healthy controls). Furthermore, it was found from both the mice model and the human serum sample study that the level of vascular endothelial growth factor (VEGF, a protein that is associated with tumor angiogenesis) adsorbed to the AuNPs is decreased in cancer samples compared to non-cancerous or less malignant cancer samples. CONCLUSION The molecular aberrations observed from this study may become new biomarkers for prostate cancer detection. The nanoparticle immunoassay reported here can be used as a convenient and general tool to screen and analyze serum proteins and to discover new biomarkers associated with cancer and other human diseases.
Collapse
Affiliation(s)
- Qun Huo
- NanoScience Technology Center and Department of Chemistry, University of Central Florida, Orlando, FL 32826, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Asmane I, Céraline J, Duclos B, Rob L, Litique V, Barthélémy P, Bergerat JP, Dufour P, Kurtz JE. New strategies for medical management of castration-resistant prostate cancer. Oncology 2011; 80:1-11. [PMID: 21577012 DOI: 10.1159/000323495] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 11/26/2010] [Indexed: 11/19/2022]
Abstract
Although advanced prostate cancer patients respond very well to front-line androgen deprivation, failure to hormonal therapy most often occurs after a median time of 18-24 months. The care of castration-resistant prostate cancer (CRPC) has significantly evolved over the past decade, with the onset of first-line therapy with docetaxel. Although numerous therapy schedules have been investigated alongside docetaxel, in either first-line or salvage therapy, results were dismal. However, CRPC chemotherapy is currently evolving, with, on the one hand, new agents targeting androgen metabolism and, on the other hand, significant progress in chemotherapy drugs, particularly for second-line therapy. The aim of the present review is to describe the current treatments for CRPC chemotherapy alongside their challengers that might shortly become new standards. In this article, we discuss the most recent data from clinical trials to provide the reader with a comprehensive, state-of-the-art overview of CRPC chemotherapy and hormonal therapy.
Collapse
Affiliation(s)
- Irène Asmane
- Department of Hematology and Oncology, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hensel JA, Chanda D, Kumar S, Sawant A, Grizzle WE, Siegal GP, Ponnazhagan S. LL-37 as a therapeutic target for late stage prostate cancer. Prostate 2011; 71:659-70. [PMID: 20957672 PMCID: PMC3025071 DOI: 10.1002/pros.21282] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 08/31/2010] [Indexed: 11/12/2022]
Abstract
BACKGROUND The antimicrobial peptide, leucine-leucine-37 (LL-37), stimulates proliferation, angiogenesis, and cellular migration, inhibits apoptosis and is associated with inflammation. Since these functional processes are often exaggerated in cancer, the aim of the present study was to investigate the expression and role of LL-37 in prostate cancer (PCa) and establish its value as a therapeutic target. METHODS We evaluated the expression of LL-37 and the murine orthologue, cathelicidin-related antimicrobial peptide (CRAMP) in human and murine prostate tumors, respectively. Compared to normal/benign prostate tissue, both LL-37 and CRAMP were increasingly over-expressed with advancing grades of primary PCa and its metastasis in human tissues and in the transgenic adenocarcinoma mouse prostate (TRAMP) model, correspondingly. We subsequently knocked-down CRAMP in the highly tumorigenic TRAMP-C1 cell line via a RNA interference strategy to examine the importance of CRAMP on cellular proliferation, angiogenesis, invasion, apoptosis, activation of signaling pathways and tumor kinetics. RESULTS Abrogation of CRAMP expression led to decreased proliferation, invasion, type IV collagenase, and the amount of phosphorylated Erk1/2 and Akt signaling in vitro. These results were paralleled in vivo. Syngenic implantation of TRAMP-C1 cells subjected to CRAMP knock-down resulted in a decreased tumor incidence and size, and the down-regulation of pro-tumorigenic mechanisms. CONCLUSIONS CRAMP knock-down in a murine PCa model analogously demonstrated the tumorigenic contributions of LL-37 in PCa and its potential as a novel therapeutic target for the treatment of PCa and potentially, other cancers over-expressing the peptide.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Animals
- Antimicrobial Cationic Peptides/biosynthesis
- Antimicrobial Cationic Peptides/metabolism
- Cathelicidins/biosynthesis
- Cathelicidins/deficiency
- Cathelicidins/genetics
- Cathelicidins/metabolism
- Cell Growth Processes/physiology
- Cell Line, Tumor
- Gene Knockdown Techniques
- Humans
- Immunohistochemistry
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Molecular Targeted Therapy/methods
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- RNA Interference
- RNA, Neoplasm/chemistry
- RNA, Neoplasm/genetics
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Jonathan A. Hensel
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294
| | - Diptiman Chanda
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294
| | - Sanjay Kumar
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294
| | - Anandi Sawant
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294
| | - William E. Grizzle
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294
| | - Gene P. Siegal
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294
| | | |
Collapse
|
34
|
|
35
|
Pang X, Zhang L, Lai L, Chen J, Wu Y, Yi Z, Zhang J, Qu W, Aggarwal BB, Liu M. 1'-Acetoxychavicol acetate suppresses angiogenesis-mediated human prostate tumor growth by targeting VEGF-mediated Src-FAK-Rho GTPase-signaling pathway. Carcinogenesis 2011; 32:904-12. [PMID: 21427164 DOI: 10.1093/carcin/bgr052] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Cancer therapeutic agents that are safe, effective and affordable are urgently needed. We describe that 1'-acetoxychavicol acetate (ACA), a component of Siamese ginger (Languas galanga), can suppress prostate tumor growth by largely abrogating angiogenesis. ACA suppressed vascular endothelial growth factor (VEGF)-induced proliferation, migration, adhesion and tubulogenesis of primary cultured human umbilical vascular endothelial cells (HUVECs) in a dose-dependent manner. ACA also inhibited VEGF-induced microvessel sprouting from aortic rings ex vivo and suppressed new vasculature formation in Matrigel plugs in vivo. We further demonstrated that the mechanisms of this chavicol were to block the activation of VEGF-mediated Src kinase, focal adhesion kinase (FAK) and Rho family of small guanosine triphosphatases (GTPases) (Rac1 and Cdc42 but not RhoA) in HUVECs. Furthermore, treatment of human prostate cancer cells (PC-3) with ACA resulted in decreased cell viability and suppression of angiogenic factor production by interference with dual Src/FAK kinases. After subcutaneous administration to mice bearing human prostate cancer PC-3 xenografts, ACA (6 mg/kg/day) remarkably inhibited tumor volume and tumor weight and decreased levels of Src, CD31, VEGF and Ki-67. As indicated by immunohistochemistry and TUNEL analysis, microvessel density and cell proliferation were also dramatically suppressed in tumors from ACA-treated mice. Taken together, our findings suggest that ACA targets the Src-FAK-Rho GTPase pathway, leading to the suppression of prostate tumor angiogenesis and growth.
Collapse
Affiliation(s)
- Xiufeng Pang
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Davies S, Dai D, Pickett G, Thiel KW, Korovkina VP, Leslie KK. Effects of bevacizumab in mouse model of endometrial cancer: Defining the molecular basis for resistance. Oncol Rep 2011; 25:855-62. [PMID: 21240464 PMCID: PMC3433042 DOI: 10.3892/or.2011.1147] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 11/22/2010] [Indexed: 11/05/2022] Open
Abstract
Endometrial cancer is the most frequent gynecologic cancer in women. Long-term outcomes for patients with advanced stage or recurrent disease are poor. Targeted molecular therapy against the vascular endothelial growth factor (VEGF) and its receptors constitute a new therapeutic option for these patients. The goal of our study was to assess the potential effectiveness of inhibition of VEGF/VEGFR signaling in a xenograft model of endometrial cancer using bevacizumab (Avastin, a humanized antibody against VEGFA). We also aimed to identify molecular markers of sensitivity or resistance to this agent. We show that bevacizumab retards tumor growth in athymic mice by inhibiting molecular components of signaling pathways that sustain cell survival and proliferation. We also demonstrate that resistance to bevacizumab may involve up-regulation of anti-apoptotic genes and certain proto-oncogenes. We propose that down-regulation of ARHGAP6 and MMP15 transcripts indicates that tumors are sensitive to bevacizumab whereas inhibition of PKCδ- or S6K-dependent signaling and up-regulation of TNFRS4 or MMP13 and MMP14 mark a developing resistance to bevacizumab therapy. Interestingly, the significant activation of c-Jun oncogene detected in bevacizumab-treated tumors suggests that, in endometrial cancers, the c-Jun-mediated pathway(s) contribute to bevacizumab resistance.
Collapse
Affiliation(s)
- Suzy Davies
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | | | | | | | | | | |
Collapse
|
37
|
Properties and effects of a novel liquid crystal nanoparticle formulation of docetaxel in a prostate cancer mouse model. Eur J Pharm Sci 2010; 41:369-75. [DOI: 10.1016/j.ejps.2010.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 07/01/2010] [Accepted: 07/04/2010] [Indexed: 11/18/2022]
|
38
|
Rosenthal SA, Sandler HM. Treatment strategies for high-risk locally advanced prostate cancer. Nat Rev Urol 2010; 7:31-8. [PMID: 20062072 DOI: 10.1038/nrurol.2009.237] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
High-risk prostate cancer can be defined by the assessment of pretreatment prognostic factors such as clinical stage, Gleason score, and PSA level. High-risk features include PSA >20 ng/ml, Gleason score 8-10, and stage T3 tumors. Patients with adverse prognostic factors have historically fared poorly with monotherapeutic approaches. Multimodal treatment utilizing combined androgen suppression and radiotherapy has improved survival rates for patients with high-risk prostate cancer. In addition, multiple randomized trials in patients treated with primary radical prostatectomy have demonstrated improved outcomes with the addition of adjuvant radiotherapy. Improved radiotherapy techniques that allow for dose escalation, and new systemic therapy approaches such as adjuvant chemotherapy, present promising future therapeutic alternatives for patients with high-risk prostate cancer.
Collapse
Affiliation(s)
- Seth A Rosenthal
- Radiation Oncology Centers, Radiological Associates of Sacramento, 1500 Expo Parkway, Sacramento, CA 95815, USA.
| | | |
Collapse
|
39
|
Open-label, phase I dose-escalation study of sodium selenate, a novel activator of PP2A, in patients with castration-resistant prostate cancer. Br J Cancer 2010; 103:462-8. [PMID: 20648008 PMCID: PMC2939789 DOI: 10.1038/sj.bjc.6605798] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background: Angiogenesis is fundamental to the progression of many solid tumours including prostate cancer. Sodium selenate is a small, water-soluble, orally bioavailable activator of PP2A phosphatase with anti-angiogenic properties. Methods: This was a dose-escalation phase I study in men with asymptomatic, chemotherapy-naïve, castration-resistant prostate cancer. The primary objective was to determine the maximum tolerated dose (MTD). Secondary objectives included establishing the safety, tolerability and pharmacokinetic profile. Results: A total of 19 patients were enrolled. The MTD was 60 mg per day. Dose-limiting toxicity (fatigue and diarrhoea) was observed at 90 mg per day. The most frequently reported treatment-related adverse events across all treatment cohorts were nausea, diarrhoea, fatigue, muscle spasms, alopecia and nail disorders. No grade 4 toxicities were observed and there were no deaths on study. Linear pharmacokinetics was observed. One patient had a PSA response >50%. Median time to PSA progression (for non-responders) was 14.2 weeks. Mean PSA doubling time increased during the main treatment phase from 2.18 months before trial to 3.85 months. Conclusion: Sodium selenate is well tolerated at a dose of 60 mg per day with modest single-agent efficacy similar to other anti-angiogenic agents. Further trials in combination with conventional cytotoxic regimens are warranted.
Collapse
|
40
|
Staibano S, Mascolo M, Di Benedetto M, Vecchione ML, Ilardi G, Di Lorenzo G, Autorino R, Salerno V, Morena A, Rocco A, Turco MC, Morelli E. BAG3 protein delocalisation in prostate carcinoma. Tumour Biol 2010; 31:461-9. [PMID: 20535599 DOI: 10.1007/s13277-010-0055-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Accepted: 05/08/2010] [Indexed: 12/01/2022] Open
Abstract
Despite the progressive increase of early diagnosis, a subset of prostate cancers show a metastasizing and lethal course, not always predictable upon the traditional prognostic parameters. The object of this study was to investigate the role of the survival co-chaperone protein BAG3 as a new prognostic marker for prostate cancer. BAG3 was detected by immunohistochemistry in 55 specimens of surgically removed prostate carcinomas and in 15 surgical specimens of non-neoplastic prostate tissues. Results were compared with clinic-pathological data and outcome of patients and statistically evaluated. BAG3 resulted expressed in all the cases: Non-neoplastic prostate tissue showed a cytoplasmatic staining with apical reinforcement, a finding which appears consistent with the reported connection of the protein with the membrane focal cell-adhesion complexes. In prostate carcinomas, BAG3 showed a progressive decrease of the expression level from well- to low-differentiated carcinoma, coupled with the loss of polarisation of the signal in metastasizing cases. These results indicate that BAG3 intra-cytoplasmic delocalisation is a specific feature of cancer versus non-neoplastic prostate and a candidate new marker for prediction of prostate cancer invasiveness and behaviour.
Collapse
Affiliation(s)
- Stefania Staibano
- Department of Biomorphological and Functional Sciences, Pathology Section, University Federico II of Naples, School of Medicine, Via S. Pansini, 5, 80131, Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Schrijvers D, Van Erps P, Cortvriend J. Castration-refractory prostate cancer: New drugs in the pipeline. Adv Ther 2010; 27:285-96. [PMID: 20532721 DOI: 10.1007/s12325-010-0038-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Indexed: 02/07/2023]
Abstract
The standard treatment for patients with castration-refractory prostate cancer (CRPC) is the combination docetaxel-prednisone, if the patient can support chemotherapy. Several new treatments have been tested in chemotherapy-naïve or docetaxel-pretreated patients with CRPC. Some of these treatments have shown activity in first-line and second-line treatment. In this review, an update is given of new treatment studies performed in patients with CRPC.
Collapse
|
42
|
Fizazi K, Sternberg CN, Fitzpatrick JM, Watson RW, Tabesh M. Role of targeted therapy in the treatment of advanced prostate cancer. BJU Int 2010; 105:748-67. [DOI: 10.1111/j.1464-410x.2010.09236.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|