Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Prostate ; 78(2): 128-139, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29131367

ABSTRACT

BACKGROUND: Nerves are key factors in prostate cancer (PCa), but the functional role of innervation in prostate cancer is poorly understood. PCa induced neurogenesis and perineural invasion (PNI), are associated with aggressive disease. METHOD: We denervated rodent prostates chemically and physically, before orthotopically implanting cancer cells. We also performed a human neoadjuvant clinical trial using botulinum toxin type A (Botox) and saline in the same patient, before prostatectomy. RESULT: Bilateral denervation resulted in reduced tumor incidence and size in mice. Botox treatment in humans resulted in increased apoptosis of cancer cells in the Botox treated side. A similar denervation gene array profile was identified in tumors arising in denervated rodent prostates, in spinal cord injury patients and in the Botox treated side of patients. Denervation induced exhibited a signature gene profile, indicating translation and bioenergetic shutdown. Nerves also regulate basic cellular functions of non-neoplastic epithelial cells. CONCLUSION: Nerves play a role in the homeostasis of normal epithelial tissues and are involved in prostate cancer tumor survival. This study confirms that interactions between human cancer and nerves are essential to disease progression. This work may make a major impact in general cancer treatment strategies, as nerve/cancer interactions are likely important in other cancers as well. Targeting the neural microenvironment may represent a therapeutic approach for the treatment of human prostate cancer.


Subject(s)
Botulinum Toxins, Type A/pharmacology , Denervation/methods , Prostate , Prostatic Neoplasms , Acetylcholine Release Inhibitors/pharmacology , Animals , Disease Models, Animal , Disease Progression , Energy Metabolism , Male , Mice , Neoplasm Invasiveness , Prostate/innervation , Prostate/pathology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Tumor Burden , Tumor Microenvironment/physiology
2.
Invest New Drugs ; 32(2): 235-242, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23609828

ABSTRACT

BACKGROUND: Given the immunogenicity of NY-ESO-1 peptides in prostate cancer, a phase I clinical trial was designed to evaluate HLA class-I and class-II restricted NY-ESO-1 peptides in metastatic castration-resistant prostate cancer (mCRPC). METHODS: Patients with progressive mCRPC, Zubrod Performance Status ≤2, PSA ≥10 ng/ml who had appropriate HLA class I (A2) and class II haplotypes (DR4, DP4) were eligible. Three groups with 3 patients each received the vaccine subcutaneously every 2 weeks for 6 doses. Group 1 received a peptide presented by an HLA class I haplotype (HLA-A2), Group 2 with a peptide presented by HLA class II haplotype (DR4, DP4), and Group 3 with peptides presented by both Class I and II haplotypes. Androgen-deprivation was continued. Owing to a myocardial infarction, the protocol was amended to omit the use of GM-CSF. RESULTS: Fourteen patients were evaluable for toxicities and 9 received all 6 doses and were evaluable for efficacy. One death from myocardial infarction following GM-CSF occurred in a patient with generalized myalgias. After omitting GM-CSF, no grade >2 toxicities were observed. Among 9 patients evaluable for efficacy, the median PSA doubling time pre-therapy and during therapy were 3.1 and 4.92 months, respectively. NY-ESO-1 specific T-cell response observed by ELISPOT appeared more frequent in docetaxel-naïve patients (4 of 4) than docetaxel-pretreated patients (2 of 5). CONCLUSION: In men with mCRPC, individualized HLA class-I and/or class-II restricted NY-ESO-1 peptides were tolerable, appeared to slow PSA doubling time and yielded antigen-specific T-cell responses more often in chemonaïve patients.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines , Immunotherapy , Membrane Proteins/immunology , Peptides/therapeutic use , Prostatic Neoplasms, Castration-Resistant/therapy , Aged , Aged, 80 and over , HLA Antigens , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Prostatic Neoplasms, Castration-Resistant/immunology , T-Lymphocytes/immunology
3.
Eur Urol Focus ; 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38242825

ABSTRACT

CONTEXT: Some authors propose extended pelvic lymph node dissection (ePLND) to enhance diagnostic and therapeutic outcomes in patients with localized prostate cancer. However, recent evidence found no difference in biochemical recurrence (BCR). OBJECTIVE: To stratify and analyze available evidence on ePLND and its impact on BCR in patients with localized prostate cancer. EVIDENCE ACQUISITION: We systematically reviewed the literature according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines to identify studies up to November 2023. We identified original articles that presented statistical comparisons through Cox regressions reported as hazard ratio (HR) or survival curve data reported as Kaplan-Meier curve differences in BCR in patients undergoing radical prostatectomy and stratified by the extent of lymph node dissection for localized prostate cancer. EVIDENCE SYNTHESIS: We identified 12 studies, with two being randomized controlled trials (RCTs). The RCTs showed no benefit of ePLND with an HR of 1.03 ([0.92, 1.14], p = 0.61). A combined analysis with the ten retrospective studies revealed a notable reduction in BCR with an HR of 0.68 ([0.52, 0.88], p = 0.003). A subgroup analysis based on the extent of dissection demonstrated that studies focusing on the more conservative extended template of dissection did not show significant BCR benefit (HR 0.97 [0.72, 1.32], p = 0.86). In contrast, dissections that expanded the anatomical extent showed decreased BCR (HR 0.56 [0.41, 0.75], p < 0.0001). A Bayesian network analysis highlights significant differences in BCR reduction between different dissection approaches, indicating the potential benefits of specific dissection templates. CONCLUSIONS: Available literature on the extent of pelvic lymph node dissection needs to be improved in quality and varying definitions of the ePLND template. Dissection of the common iliac nodes may be beneficial. PATIENT SUMMARY: There is a potential benefit in removing more lymph nodes during radical prostatectomy. However, more research is needed to determine whether this strategy benefits certain patient groups.

4.
J Robot Surg ; 18(1): 177, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38630430

ABSTRACT

Lymphocele is one of the most common complications after radical prostatectomy. Multiple authors have proposed the use of vessel sealants or peritoneal interposition techniques as preventive interventions. This study aimed to aggregate and analyze the available literature on different interventions which seek to prevent lymphocele through a Bayesian Network. A systematic review was performed to identify prospective studies evaluating strategies for lymphocele prevention after robot assisted laparoscopic prostatectomy + pelvic lymph node dissection. Data was inputted into Review Manager 5.4 for pairwise meta-analysis. Data was then used to build a network in R Studio. These networks were used to model 200,000 Markov Chains via MonteCarlo sampling. The results are expressed as odds ratios (OR) with 95% credible intervals (CrI). Meta-regression was used to determine coefficient of change and adjust for pelvic lymph node dissection extent. Ten studies providing data from 2211 patients were included. 1097 patients received an intervention and 1114 patients served as controls. Interposition with fenestration had the lowest risk of developing a lymphocele (OR 0.14 [0.04, 0.50], p = 0.003). All interventions, except sealants or patches, had significant decreased odds of lymphocele rates. Meta-analysis of all the included studies showed a decreased risk of developing a lymphocele (OR 0.42 [0.33, 0.53], p < 0.00001) for the intervention group. Perivesical fixation and interposition with fenestration appear to be effective interventions for reducing the overall incidence of lymphocele.


Subject(s)
Lymphocele , Robotic Surgical Procedures , Humans , Male , Bayes Theorem , Lymph Node Excision/adverse effects , Lymphocele/etiology , Lymphocele/prevention & control , Network Meta-Analysis , Prospective Studies , Prostatectomy/adverse effects , Robotic Surgical Procedures/methods
5.
Am J Pathol ; 178(1): 79-87, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224046

ABSTRACT

We previously reported that reactive stroma grading in prostate cancer (PCa) is predictive of biochemical recurrence in prostatectomies and biopsies. In this study, we tested whether quantifying the percentage of reactive stromal grade 3 (RSG 3; stromogenic carcinoma pattern) in the entire tumor is predictive of PCa-specific death. Whole-mount prostatectomies operated by a single surgeon obtained between 1983 and 1998 were reviewed. Reactive stroma was evaluated as described previously, and areas of RSG 3 in the entire tumor were registered as percentages of total tumor. Statistical analysis was performed using Spearman, Kaplan-Meier, and Cox analyses. In all, 872 cases were evaluable. Quantification of RSG 3 percentage was an independent predictor of biochemical recurrence, analyzed as a continuous or grouped variable. Patients with higher RSG 3 percentages (larger tumor areas with RSG 3) had a significantly decreased biochemical recurrence-free survival than those with a lower RSG 3 percentage, even within the Gleason score 7 subset of patients. A nomogram introduced this new variable to the model. Furthermore, quantification of RSG 3 percentage was significantly predictive of PCa-specific death. Quantification of the RSG 3 (stromogenic carcinoma) area in PCa provides additional novel information on prognosis. These data substantiate the concept that the tumor microenvironment holds significant predictive information, as well as biological significance.


Subject(s)
Carcinoma/mortality , Carcinoma/pathology , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Aged , Carcinoma/surgery , Cohort Studies , Disease-Free Survival , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Prostatectomy , Prostatic Neoplasms/surgery
6.
Clin Cancer Res ; 14(23): 7593-603, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047084

ABSTRACT

PURPOSE: Perineural invasion is the only interaction between cancer cells and nerves studied to date. It is a symbiotic relationship between cancer and nerves that results in growth advantage for both. In this article, we present data on a novel biological phenomenon, cancer-related axonogenesis and neurogenesis. EXPERIMENTAL DESIGN: We identify spatial and temporal associations between increased nerve density and preneoplastic and neoplastic lesions of the human prostate. RESULTS: Nerve density was increased in cancer areas as well as in preneoplastic lesions compared with controls. Two- and three-dimensional reconstructions of entire prostates confirmed axonogenesis in human tumors. Furthermore, patients with prostate cancer had increased numbers of neurons in their prostatic ganglia compared with controls, corroborating neurogenesis. Finally, two in vitro models confirmed that cancer cells, particularly when interacting with nerves in perineural invasion, induce neurite outgrowth in prostate cancer. Neurogenesis is correlated with features of aggressive prostate cancer and with recurrence in prostate cancer. We also present a putative regulatory mechanism based on semaphorin 4F (S4F). S4F is overexpressed in cancers cells in the perineural in vitro model. Overexpression of S4F in prostate cancer cells induces neurogenesis in the N1E-115 neurogenesis assay and S4F inhibition by small interfering RNA blocks this effect. CONCLUSIONS: This is the first description of cancer-related neurogenesis and its putative regulatory mechanism.


Subject(s)
Neoplasm Invasiveness/pathology , Neurogenesis/physiology , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Humans , Male , Peripheral Nerves/pathology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Prostate/innervation , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Semaphorins/metabolism , Tissue Array Analysis
7.
Clin Cancer Res ; 14(22): 7511-8, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010869

ABSTRACT

PURPOSE: To assess the safety of administering bortezomib to patients undergoing a radical prostatectomy, to assess pathologic changes induced by bortezomib in prostate cancer specimen, and to verify alterations by the drug in proteasome protein targets. EXPERIMENTAL DESIGN: Bortezomib is a proteasome inhibitor that has shown activity in vitro and in vivo in prostate cancer. We performed a neoadjuvant clinical trial of bortezomib in men with prostate cancer at high risk of recurrence. The primary endpoints were to evaluate safety and biological activity. RESULTS: Bortezomib is generally safe in the preoperative setting. Antitumor activity was manifested by tumor cytopathic effect, drops in serum prostate-specific antigen in some patients, and increases in tumor apoptosis. This was associated with cytoplasmic entrapment of nuclear factor-kappaB. We found an unexpected increase in proliferation in treated tissues and in vitro. Bortezomib also increased SRC-3 levels and phosphorylated Akt, both in vitro and in treated prostate cancer tissues. Knockdown of SRC-3 blocked the increase in activated Akt in vitro. Combined treatment with bortezomib and the Akt inhibitor perifosine was more effective than either agent alone in vitro. CONCLUSION: These data suggest that combined therapies targeting the proteasome and the Akt pathway may have increased efficacy.


Subject(s)
Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Histone Acetyltransferases/drug effects , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/drug effects , Pyrazines/therapeutic use , Trans-Activators/drug effects , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Blotting, Western , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Flow Cytometry , Histone Acetyltransferases/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , NF-kappa B/drug effects , Neoadjuvant Therapy , Nuclear Receptor Coactivator 3 , Phosphorylcholine/administration & dosage , Phosphorylcholine/analogs & derivatives , Prostate-Specific Antigen/blood , Prostate-Specific Antigen/drug effects , Prostatectomy , Prostatic Neoplasms/surgery , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Trans-Activators/metabolism
8.
Hum Pathol ; 39(2): 282-91, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18206496

ABSTRACT

We previously reported that reactive stromal grading in radical prostatectomies is a predictor of recurrence and that reactive stromal grading 0 and 3 are associated with lower biochemical recurrence-free survival rates than reactive stromal grading 1 and 2. We explored the prognostic significance of reactive stromal grading in preoperative needle biopsies. At Baylor College of Medicine, 224 cases of prostatic carcinoma were diagnosed by needle biopsy. Reactive stromal grading was evaluated on hematoxylin-eosin (H&E)-stained sections on the basis of previously described criteria: grade 0, with 0% to 5% reactive stroma; grade 1, 6% to 15%; grade 2, 16% to 50%; grade 3, 51% to 100%, or at least a 1:1 ratio between glands and stroma. Kaplan-Meier and Cox proportional hazard analyses were used. Reactive stromal grading distribution was as follows: reactive stromal grading 0, 1 case (0.5%); reactive stromal grading 1, 149 cases (66.5%); reactive stromal grading 2, 59 cases (26.3%); reactive stromal grading 3, 15 cases (6.7%). Reactive stromal grading in biopsies was correlated with adverse clinicopathologic parameters in the prostatectomy. Patients with reactive stromal grading 1 and 2 had better survival than those with 0 and 3 (P = .0034). Reactive stromal grading was an independent predictor of recurrence (hazard ratio = 1.953; P = .0174). Reactive stromal grading is independent of Gleason 4 + 3 and 3 + 4 in patients with a Gleason score of 7. Quantitation of reactive stroma and recognition of the stromogenic carcinoma in H&E-stained biopsies is useful to predict biochemical recurrence in prostate carcinoma patients independent of Gleason grade and prostate-specific antigen.

9.
Hum Pathol ; 38(11): 1688-95, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17707459

ABSTRACT

Up-regulation of caveolin-1 (cav-1) has been implicated in human prostate cancer progression/metastasis and shown to promote cancer cell survival. It has also been shown that cav-1 is secreted by tumor cells and may regulate the growth, functional activities, and migration of vascular endothelial cells. However, the relationship of cav-1 expression in prostate cancer cells and tumor associated endothelial cells (TAEC) to tumor-associated angiogenesis remains to be investigated. Dual immunofluorescent labeling with antibodies to CD34 and cav-1 was performed on 56 prostate cancer specimens obtained by radical prostatectomy and stratified according to cav-1 positivity in cancer cells. The tumor microvessel densities (MVD) and cav-1 expression in TAEC within these specimens were measured and correlated with cav-1 expression in prostate cancer cells. The MVD values were significantly higher in cav-1-positive (n = 25) than in the cav-1-negative (n = 31) tumors (median of 44 versus 25 vessels/field, P = .0140). Additional studies showed that the cav-1 positivity in microvessels within tumor specimens was significantly less frequent than in the blood vessels of benign prostatic tissues (94.4% versus 98.6%, P = .0012). In contrast, the percentage of cav-1-positive TAEC in cav-1-positive tumors was significantly higher than in cav-1-negative tumors (95.8% versus 92.7%, P = .0024). This increased cav-1 positivity in TAEC was predominantly confined to regions with cav-1-positive tumor cells corresponding to the higher percentage of cav-1-positive microvessels within these regions in cav-1-positive, as opposed to cav-1-negative tumors (P = .0086). These positive correlations provide new evidence for the involvement of prostate cancer cell derived cav-1 in mediating angiogenesis during prostate cancer progression. They also establish a conceptual framework for further investigation of cav-1 proangiogenic activities.


Subject(s)
Caveolin 1/physiology , Neovascularization, Pathologic/physiopathology , Prostatic Neoplasms/metabolism , Endothelial Cells/physiology , Humans , Male , Prostatic Neoplasms/pathology , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Hum Pathol ; 38(11): 1611-20, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17868773

ABSTRACT

We previously reported that reactive stromal grading in radical prostatectomies is a predictor of recurrence and that reactive stromal grading 0 and 3 are associated with lower biochemical recurrence-free survival rates than reactive stromal grading 1 and 2. We explored the prognostic significance of reactive stromal grading in preoperative needle biopsies. At Baylor College of Medicine, 224 cases of prostatic carcinoma were diagnosed by needle biopsy. Reactive stromal grading was evaluated on hematoxylin-eosin (H&E)-stained sections on the basis of previously described criteria: grade 0, with 0% to 5% reactive stroma; grade 1, 6% to 15%; grade 2, 16% to 50%; grade 3, 51% to 100%, or at least a 1:1 ratio between glands and stroma. Kaplan-Meier and Cox proportional hazard analyses were used. Reactive stromal grading distribution was as follows: reactive stromal grading 0, 1 case (0.5%); reactive stromal grading 1, 149 cases (66.5%); reactive stromal grading 2, 59 cases (26.3%); reactive stromal grading 3, 15 cases (6.7%). Reactive stromal grading in biopsies was correlated with adverse clinicopathologic parameters in the prostatectomy. Patients with reactive stromal grading 1 and 2 had better survival than those with 0 and 3 (P = .0034). Reactive stromal grading was an independent predictor of recurrence (hazard ratio = 1.953; P = .0174). Reactive stromal grading is independent of Gleason 4 + 3 and 3 + 4 in patients with a Gleason score of 7. Quantitation of reactive stroma and recognition of the stromogenic carcinoma in H&E-stained biopsies is useful to predict biochemical recurrence in prostate carcinoma patients independent of Gleason grade and prostate-specific antigen.


Subject(s)
Prostatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Biopsy, Needle , Cohort Studies , Disease-Free Survival , Humans , Male , Middle Aged , Predictive Value of Tests , Prognosis , Prostate/pathology , Prostatectomy , Prostatic Neoplasms/surgery , Recurrence , Stromal Cells/pathology
11.
Clin Cancer Res ; 12(16): 4872-5, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16914574

ABSTRACT

PURPOSE: Up-regulation of caveolin-1 (cav-1) is associated with virulent prostate cancer, and serum cav-1 levels are elevated in prostate cancer patients but not in benign prostatic hyperplasia. In this study, we evaluated the potential of high preoperative serum cav-1 levels to predict biochemical progression of prostate cancer. The value of the combined preoperative markers, prostate-specific antigen (PSA), biopsy Gleason score, and serum cav-1 for predicting biochemical recurrence was also investigated. EXPERIMENTAL DESIGN: Serum samples taken from 419 prostate cancer patients before radical prostatectomy were selected from our Specialized Programs of Research Excellence prostate cancer serum and tissue bank. Serum samples were obtained 0 to 180 days before surgery and all patients had complete data on age, sex, race, stage at enrollment, and follow-up for biochemical recurrence. Serum cav-1 levels were measured according to our previously reported ELISA protocol. RESULTS: Cav-1 levels were measured in the sera of 419 prostate cancer patients; the mean serum level was 4.52 ng/mL (median 1.01 ng/mL). Patients with high serum cav-1 levels had a 2.7-fold (P = 0.0493) greater risk of developing biochemical recurrence compared with those with low serum cav-1 levels. Importantly, patients with serum PSA >/= 10 ng/mL and elevated levels of serum cav-1 had 2.44 times higher risk (P = 0.0256) of developing biochemical recurrence compared with patients with low levels of cav-1. In addition, high serum cav-1 levels combined with increasing biopsy Gleason score predicted much shorter recurrence-free survival in the group of patients with PSA >/= 10 ng/mL (P = 0.0353). Cav-1 was also able to distinguish between high- and low- risk patients with biopsy Gleason score of seven, after adjusting, for patients PSA levels (P = 0.0429). CONCLUSIONS: Overall, elevated preoperative levels of serum cav-1 predict decreased time to cancer recurrence. In the subset of patients with serum PSA of >/=10 ng/mL, the combination of serum cav-1 and biopsy Gleason score has the capacity to predict time to biochemical recurrence.


Subject(s)
Biomarkers, Tumor/blood , Caveolin 1/blood , Neoplasm Recurrence, Local/blood , Prostatic Neoplasms/blood , Adult , Aged , Cohort Studies , Humans , Male , Middle Aged , Prognosis , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/surgery
12.
Hum Pathol ; 63: 202-211, 2017 05.
Article in English | MEDLINE | ID: mdl-28315427

ABSTRACT

Prostatic carcinoma, like many other carcinomas, generates a stromal reaction. This phenomenon is well established in the scientific literature. The normal parenchymal smooth muscle phenotype switches to a myofibroblastic phenotype in response to the presence of cancer cells, with an expansion of the extracellular matrix compartment. The amount of reactive stroma is a predictor of biochemical recurrence in both radical prostatectomies and biopsies. It is a predictor of prostate cancer-specific death in prostatectomies. The aim of this study is to improve our histologic understanding of reactive stroma in prostate cancer and to determine the histologic features of the malignant epithelium found in stromogenic carcinomas or carcinomas with reactive stromal grade 3. Tissue microarrays of 800 patients and hematoxylin and eosin-stained sections of 120 radical prostatectomies, previously determined to contain a high proportion of areas with stromogenic carcinoma, were evaluated and findings systematically recorded. We identified 3 histologic patterns of reactive stroma: extracellular matrix-rich, cellular variant and edematous/myxoid variant. The most common pattern of carcinoma in stromogenic areas is of the acinar type with angulated glands and periglandular halos. The nuclei are enlarged, opened, with prominent nucleoli. Luminal borders are undulated, and amorphous pink secretion is often seen. Perineural invasion is frequently identified. Because of the clinical relevance, identification and quantification of areas with high reactive stromal grade by pathologists and reproducibility of our findings by others become essential. We believe that with the previously proposed grading system and the present morphologic description, both can be achieved.


Subject(s)
Carcinoma/pathology , Prostatic Neoplasms/pathology , Stromal Cells/pathology , Biopsy , Carcinoma/surgery , Databases, Factual , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Phenotype , Prostatectomy , Prostatic Neoplasms/surgery , Tissue Array Analysis , Tumor Microenvironment
13.
Cancer Biol Ther ; 5(1): 78-83, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16357515

ABSTRACT

Targeted therapy of proteasome regulated gene expression has potential utility in cancer treatment since components of ubiquitin-mediated proteolysis are altered in human malignancy. Specific regulators of proteasome degradation such as F-box proteins of the SCF E3 ligase complex are ideal biomarkers for assessing therapeutic efficacy since these components determine substrate specificity. An F-box protein that appears to be important in this process is human Cdc4 (Fbw7) since expression is detected in a variety of human cancers including breast, colon, pancreas and uterus. The role of Cdc4 in tumorigenesis appears to be related at least in part to regulation of cyclin E since inactivating mutations of CDC4 in cancer cells leads to cyclin E overexpression and genomic instability. In order to investigate the potential biological and clinical consequences of proteasome inhibition with respect to Cdc4 mediated targeted proteolysis, we investigated CDC4 expression and genetic alterations in 53 primary human prostate cancers in addition to correlation with relevant histopathological and clinical parameters. We identified genetic alterations in 6% of our prostate cancers while differential expression of Cdc4 isoforms correlated with advanced pathological stage and clinical recurrence. Our data suggest that CDC4 expression in prostate cancer has important biological and clinical implications since genetic alterations, differential Cdc4 isoform expression, histopathological and clinical correlation were demonstrated in our analysis. Therefore molecular genetic analysis of CDC4 expression may be an important biomarker for concurrent or subsequent clinical investigation of proteasome targeted therapy in men with prostate cancer.


Subject(s)
Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , F-Box Proteins/genetics , Gene Expression , Neoplasm Recurrence, Local/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Ubiquitin-Protein Ligases/genetics , Antineoplastic Agents/therapeutic use , F-Box-WD Repeat-Containing Protein 7 , Humans , Male , Mutation , Prostatic Neoplasms/drug therapy , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors
14.
Technol Cancer Res Treat ; 5(1): 23-36, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16417399

ABSTRACT

The advances in radiotherapy (3D-CRT, IMRT) have enabled high doses of radiation to be delivered with the least possible associated toxicity. However, the persistence of cancer (local recurrence after radiotherapy) despite these increased doses as well as distant failure suggesting the existence of micro-metastases, especially in the case of higher risk disease, have underscored the need for continued improvement in treatment strategies to manage local and micro-metastatic disease as definitively as possible. This has prompted the idea that an increase in the therapeutic index of radiotherapy might be achieved by combining it with in situ gene therapy. The goal of these combinatorial therapies is to maximize the selective pressure against cancer cell growth while minimizing treatment-associated toxicity. Major efforts utilizing different gene therapy strategies have been employed in conjunction with radiotherapy. We reviewed our and other published clinical trials utilizing this combined radio-genetherapy approach including their associated pre-clinical in vitro and in vivo models. The use of in situ gene therapy as an adjuvant to radiation therapy dramatically reduced cell viability in vitro and tumor growth in vivo. No significant worsening of the toxicities normally observed in single-modality approaches were identified in Phase I/II clinical studies. Enhancement of both local and systemic T-cell activation was noted with this combined approach suggesting anti-tumor immunity. Early clinical outcome including biochemical and biopsy data was very promising. These results demonstrate the increased therapeutic efficacy achieved by combining in situ gene therapy with radiotherapy in the management of local prostate cancer. The combined approach maximizes tumor control, both local-regional and systemic through radio-genetherapy induced cytotoxicity and anti-tumor immunity.


Subject(s)
Genetic Therapy , Prostatic Neoplasms/therapy , Animals , Clinical Trials as Topic , Combined Modality Therapy , Humans , Male , Prostatic Neoplasms/radiotherapy
15.
Int J Radiat Oncol Biol Phys ; 54(1): 101-6, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12182979

ABSTRACT

PURPOSE: The advent of widespread prostate-specific antigen screening has resulted in more younger, potent men being diagnosed with early-stage, organ-confined prostate cancer amenable to definitive surgery. Nerve-sparing prostatectomy is a relatively new surgical advance in the treatment of prostate cancer. Very few data exist on the effect of postoperative radiotherapy (RT) on erectile function after nerve-sparing prostatectomy. They are based on conventional techniques using moderate doses of radiation, 45-54 Gy. Intensity-modulated RT (IMRT) is becoming more widespread because it allows dose escalation with increased sparing of the surrounding normal tissue. We investigated the effect of postprostatectomy, high-dose IMRT on patients' erectile function. METHODS AND MATERIALS: A review of patient records found 51 patients treated between April 1998 and December 2000 with IMRT after unilateral or bilateral nerve-sparing prostatectomy. The pathologic disease stage in these patients was T2 in 47.4% and T3 in 52.6%. Postoperatively, 4 patients received hormonal ablation consisting of one injection of Lupron Depot (30 mg) 2 months before RT. The median age was 65 years (range 46-77) at the time of RT. The prescribed dose was 64 Gy (range 60-66). The mean dose was 69.6 Gy (range 64.0-72.3). Erectile function was assessed before and after RT by questionnaires. Sexual potency was defined as erectile rigidity adequate for vaginal penetration. RESULTS: Of the 51 patients, 18 (35.3%) maintained their potency and 33 (64.7%) became impotent after nerve-sparing prostatectomy. Patients who underwent bilateral nerve-sparing prostatectomy had higher rates of postoperative potency than did those who underwent unilateral nerve-sparing surgery (72.2% vs. 27.8%; p = 0.025). The follow-up for the entire group was 19.5 months. All 18 patients (100%) who were potent postoperatively remained potent after RT. The median follow-up for the 18 potent patients was 27.2 months, significantly longer than that of the impotent group, 13.0 months (p <0.001). CONCLUSION: This is the first report on the effects of dose-escalated IMRT on men who have undergone nerve-sparing prostatectomy. Despite the high dose (mean dose 69.6 Gy) to the prostate bed and nerves, postoperative IMRT had no negative effect on erectile function for the patients who remained potent after nerve-sparing prostatectomy. Longer term follow-up and a larger cohort of patients are warranted to confirm these findings.


Subject(s)
Prostatectomy/methods , Prostatic Neoplasms/radiotherapy , Aged , Follow-Up Studies , Humans , Male , Middle Aged , Penile Erection/radiation effects , Prostatectomy/adverse effects , Prostatic Neoplasms/physiopathology , Prostatic Neoplasms/surgery , Radiotherapy Dosage
16.
Int J Radiat Oncol Biol Phys ; 59(2): 562-71, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15145177

ABSTRACT

PURPOSE: In situ cytotoxic gene therapy can potentially trigger a systemic immune response, which could impact occult metastatic disease. We are currently conducting three clinical trials using in situ adenoviral vector mediated herpes simplex virus-thymidine kinase (HSV-tk) gene delivery followed by the HSV-tk prodrug ganciclovir (GCV) or valacyclovir (VCV). This study evaluates the systemic T-cell response after gene therapy in each trial. METHODS AND MATERIALS: The study protocol included three separate clinical trials in the Baylor Prostate Cancer SPORE Program: Trial A gene therapy in prostate cancer patients failing radiotherapy (36 patients), Trial B neoadjuvant gene therapy in pre-radical prostatectomy patients (22 patients), and Trial C gene therapy in combination with radiotherapy for prostate cancer (27 patients). Heparinized blood was collected at the time of vector injection and at selected intervals afterward. A complete blood count was performed, and peripheral blood lymphocytes were analyzed by fluorescent antibody cell sorting after labeling with dual color-labeled antibody pairs. RESULTS: The pretreatment mean percentage of activated CD8+ T cells (DR+CD8+ T cells) was 12.23%, 16.72%, and 14.09% (Trials A, B, and C, respectively). Two weeks posttreatment, this increased to 22.87%, 26.15%, and 39.04% (Trials A, B, and C, respectively), and these increases were statistically significant (p = 0.0188, p = 0.0010, p < 0.0001, respectively). The increase of DR+CD8+ T cells was significantly larger in Trial C than in Trial A (p = 0.0044) or Trial B (p = 0.0288). Total CD8+ T cells significantly increased at 2 weeks posttreatment in Trial B and C (p = 0.0013, p = 0.0004, respectively). Interestingly, only in Trial C were significant increases in activated CD4+ T cells seen at 2 weeks (p = 0.0035). CONCLUSIONS: This is the first report of systemic T-cell responses after HSV-tk+GCV/VCV gene therapy under three clinical trial conditions. There was an increase in activated CD8+ T cells in the peripheral blood after vector injection, suggesting the potential for activation of components of cell-mediated immune response in all trial conditions. The addition of radiotherapy to in situ gene therapy seems to further increase the total CD8+ T cells and activated CD4+ T cells.


Subject(s)
Acyclovir/analogs & derivatives , CD8-Positive T-Lymphocytes/immunology , Genetic Therapy , Lymphocyte Activation/immunology , Prostatic Neoplasms/therapy , Thymidine Kinase/genetics , Valine/analogs & derivatives , Acyclovir/therapeutic use , Adenoviridae/genetics , Adult , Aged , Aged, 80 and over , Antiviral Agents/therapeutic use , Avian Sarcoma Viruses/genetics , CD4-Positive T-Lymphocytes/immunology , Combined Modality Therapy , Ganciclovir/therapeutic use , Humans , Male , Middle Aged , Prodrugs/therapeutic use , Prostatectomy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/radiotherapy , Valacyclovir , Valine/therapeutic use
17.
Int J Radiat Oncol Biol Phys ; 58(5): 1520-9, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15050332

ABSTRACT

PURPOSE: There is an evolving role for combining radiotherapy (RT) with gene therapy in the management of prostate cancer. However, the clinical results of this combined approach are much needed. The preliminary results addressing the safety of this Phase I-II study combining RT and gene therapy (adenovirus/herpes simplex virus-thymidine kinase gene/valacyclovir with or without hormonal therapy) in the treatment of prostate cancer have been previously reported. We now report the prostate-specific antigen (PSA) response and biopsy data. METHODS AND MATERIALS: This trial was composed of three separate arms. Arm A consisted of low-risk patients (Stage T1-T2a, Gleason score <7, pretreatment PSA <10 ng/mL) treated with combined RT-gene therapy. A mean dose of 76 Gy was delivered to the prostate with intensity-modulated RT. They also received adenovirus/herpes simplex virus-thymidine kinase/valacyclovir gene therapy. Arm B consisted of high-risk patients (Stage T2b-T3, Gleason score >6, pretreatment PSA level >10 ng/mL) treated with combined RT-gene therapy and hormonal therapy (luteinizing hormone-releasing hormone agonist [30-mg Lupron, 4-month depot] and an antiandrogen [flutamide, 250 mg t.i.d. for 14 days]). Arm C consisted of patients with Stage D1 (positive pelvic lymph nodes) who received the same regimen as Arm B with the addition of 45 Gy to the pelvic lymphatics. PSA determination and biopsy were performed before, during, and after treatment. The American Society for Therapeutic Radiology and Oncology consensus definition (three consecutive rises in PSA level) was used to denote PSA failure. RESULTS: Fifty-nine patients (29 in Arm A, 26 in Arm B, and 4 in Arm C) completed the trial. The median age was 68 years (range, 39-85 years). The median follow-up for the entire group was 13.5 months (range, 1.4-27.8 months). Only Arm A patients were observed to have an increase in PSA on Day 14. The PSA then declined appropriately. All patients in Arm A (median follow-up, 13.4 months) and Arm B (median follow-up, 13.9 months) had biochemical control at last follow-up. Three patients in Arm C (with pretreatment PSA of 335, 19.6, and 2.5 ng/mL and a combined Gleason score of 8, 9, and 9 involving all biopsy cores) had biochemical failure at 3, 3, and 7.7 months. Two patients had distant failure in bone and 1 patient in the para-aortic lymph nodes outside the RT portal. Six to twelve prostate biopsies performed in these 3 patients revealed no evidence of residual carcinoma. In Arm A, biopsy showed no evidence of carcinoma in 66.7% (18 of 27), 92.3% (24 of 26), 91.7% (11 of 12), 100% (8 of 8), and 100% (6 of 6) at 6 weeks, 4 months, 12 months, 18 months, and 24 months after treatment, respectively. In Arm B, no evidence of carcinoma on biopsy was noted in 96% (24 of 25), 90.5% (19 of 21), 100% (14 of 14), 100% (7 of 7), and 100% (2 of 2), respectively, in the same interval after treatment. CONCLUSION: This is the first reported trial of its kind in the field of prostate cancer that aims to expand the therapeutic index of RT by combining it with in situ gene therapy. The initial transient PSA rise in the Arm A patients may have been a result of local immunologic response or inflammation elicited by in situ gene therapy. Additional investigation to elucidate the mechanisms is needed. Hormonal therapy may have obliterated this rise in Arm B and C patients. The biopsy data were encouraging and appeared to show no evidence of malignancy earlier than historical data. Combined RT, short-course hormonal therapy, and in situ therapy appeared to provide good locoregional control but inadequate systemic control in patients with positive pelvic lymph nodes. Longer term use of hormonal therapy in addition to gene therapy and RT has been adopted for this group of patients to maximize both locoregional and systemic control.


Subject(s)
Acyclovir/analogs & derivatives , Genetic Therapy/methods , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/therapy , Radiotherapy, Conformal , Thymidine Kinase/therapeutic use , Valine/analogs & derivatives , Acyclovir/therapeutic use , Adenoviridae/genetics , Adult , Aged , Aged, 80 and over , Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Antiviral Agents/therapeutic use , Biopsy , Combined Modality Therapy , Flutamide/therapeutic use , Follow-Up Studies , Genetic Vectors/therapeutic use , Humans , Leuprolide/therapeutic use , Male , Middle Aged , Prodrugs/therapeutic use , Prostate/pathology , Prostatic Neoplasms/pathology , Radiotherapy Dosage , Thymidine Kinase/genetics , Valacyclovir , Valine/therapeutic use
18.
Curr Drug Targets ; 4(3): 251-61, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12643475

ABSTRACT

Prostate cancer is the most commonly diagnosed non-cutaneous cancer in adult males. Although prostate cancer that is confined to the gland can be cured in many patients using surgery or radiation, these treatments are only effective for localized tumors and the long-term failure rates for these treatments suggests that prostate cancer can metastasize relatively early in the course of the disease. Once prostate cancer has metastasized there are no curative therapies. The greatest challenge in the treatment of advanced prostate cancer is to access and eliminate metastatic cells. Therefore, effective prostate cancer therapy will require novel strategies to target cancer cells both at the site of the primary tumor and at distant metastatic sites. In this article we review several therapeutic targets and approaches that may provide new treatments for metastatic prostate cancer. We discuss the use of small molecules to target specific molecular events associated with metastatic prostate cancer, the use of specific antibodies that target unique metastasis associated molecules and the use of various gene therapy strategies to achieve anti-metastatic activities.


Subject(s)
Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Animals , Antibodies/chemistry , Antibodies/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/immunology , Cancer Vaccines/therapeutic use , Genetic Therapy/methods , Humans , Immunotherapy , Male , Neoplasm Metastasis , Prostatic Neoplasms/immunology
19.
Expert Rev Anticancer Ther ; 2(3): 309-21, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12113054

ABSTRACT

Prostate cancer is the most common noncutaneous cancer in man. When confined to the prostate it can be cured by radical prostatectomy or irradiation therapy. However, there are no curative therapies for locally advanced, recurrent or metastatic disease. Prostate cancer gene therapy has recently transition from preclinical studies to clinical trials with the goal of developing novel treatments for prostate cancer. The greatest challenge in treating advanced prostate cancer is therapeutic access to and the elimination of metastases. This review details two aspects of prostate cancer gene therapy, the types of delivery systems under development and specific categories of therapeutic genes available with an emphasis on the mechanism of action of specific gene therapy strategies.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Genes, Tumor Suppressor , Genetic Therapy/methods , Prostatic Neoplasms/therapy , Adjuvants, Immunologic/genetics , Apoptosis/genetics , Bacteria/enzymology , Bacteria/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/therapeutic use , Cytosine Deaminase , Diphtheria Toxin/genetics , Diphtheria Toxin/therapeutic use , Genetic Vectors , Humans , Male , Neoplasm Metastasis , Neoplasm Recurrence, Local , Nucleoside Deaminases/genetics , Nucleoside Deaminases/therapeutic use , Prodrugs/metabolism , Prodrugs/therapeutic use , Prostatic Neoplasms/genetics , Simplexvirus/enzymology , Simplexvirus/genetics , Thymidine Kinase/genetics , Thymidine Kinase/therapeutic use
20.
Clin Cancer Res ; 19(22): 6101-11, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24097862

ABSTRACT

BACKGROUND: Semaphorin 4F (S4F) has roles in embryologic axon guidance and is expressed in adults. S4F is involved in cancer-induced neurogenesis. METHODS: Prostate cells were transfected with S4F retrovirus. Cells and controls were used for a bromodeoxyuridine (BrdUrd) incorporation assay (proliferation) and in vitro scratch and Matrigel Transwell chamber invasion assay (migration). Monoclonal antibodies were developed using baculovirus-expressed recombinant GST-S4F and used to immunostain tissue microarrays. Slides were imaged using deconvolution and analyzed using tissue segmentation. Data were correlated with clinicopathologic parameters, other biomarkers and survival analysis conducted. Heterogeneity of S4F expression was analyzed with unsupervised clustering algorithms. RESULTS: Proliferation rates measured by BrdUrd incorporation were higher in all S4F-transfected cells. S4F overexpression was associated with increased motility of the cancer cells. S4F expression was overexpressed in high-grade prostatic intraepithelial neoplasia/prostate cancer than normal epithelium. S4F expression correlated with seminal vesicle invasion. Patients with high values of S4F in prostate cancer cytoplasm are at significantly higher risk of biochemical recurrence, by univariate and multivariate analyses. S4F cytoplasmic expression in prostate cancer cells also correlates with nerve density in prostate cancer and perineural invasion diameter. Correlations were identified with NF-κB and inversely with apoptosis in perineural invasion. CONCLUSION: These data show that S4F is significantly involved in human prostate cancer progression. S4F is a key regulator of the interactions between nerves in the tumor microenvironment and cancer cells. Because of the importance of cancer nerve interaction in the biology of cancer and its clinical implication, S4F can be considered a major therapeutic target. Clin Cancer Res; 19(22); 6101-11. ©2013 AACR.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuroepithelial Cells/metabolism , Neurogenesis/genetics , Prostatic Neoplasms/genetics , Semaphorins/metabolism , Apoptosis/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Male , Membrane Proteins/genetics , NF-kappa B/genetics , Neoplasm Invasiveness/genetics , Nerve Tissue Proteins/genetics , Seminal Vesicles/cytology , Seminal Vesicles/pathology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL