Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 185
Filter
1.
Ann Oncol ; 31(7): 930-941, 2020 07.
Article in English | MEDLINE | ID: mdl-32289380

ABSTRACT

BACKGROUND: We have shown previously in multivariable analysis that black men had 19% lower risk of death than white men with metastatic castration-resistant prostate cancer (mCRPC) treated with a docetaxel and prednisone (DP)-based regimen. The primary goal of this analysis was to compare progression-free survival (PFS), biochemical PFS, ≥50% decline in prostate-specific antigen (PSA) from baseline and objective response rate (ORR) in white, black and Asian men with mCRPC treated with a DP-based regimen. PATIENTS AND METHODS: Individual patient data from 8820 mCRPC men randomized on nine phase III trials to a DP-containing regimen were combined. Race used in the analysis was based on self-report. End points were PFS, biochemical PSA, ≥50% decline in PSA from baseline and ORR. The proportional hazards and the logistic regression models were employed to assess the prognostic importance of race in predicting outcomes adjusting for established prognostic factors. RESULTS: Of 8820 patients, 7528 (85%) were white, 500 (6%) were black, 424 were Asian (5%) and 368 (4%) had race unspecified. Median PFS were 8.3 [95% confidence interval (CI) 8.2-8.5], 8.2 (95% CI 7.4-8.8) and 8.3 (95% CI 7.6-8.8) months in white, black and Asian men, respectively. Median PSA PFS were 9.9 (95% CI 9.7-10.4), 8.5 (95% CI 8.0-10.3) and 11.1 (95% CI 9.9-12.5) months in white, black and Asian men, respectively. CONCLUSIONS: We observed no differences in clinical outcomes by race and ethnic groups in men with mCRPC enrolled on these phase III clinical trials with DP.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Disease-Free Survival , Docetaxel/therapeutic use , Ethnicity , Humans , Male , Prednisone/therapeutic use , Prostate-Specific Antigen , Prostatic Neoplasms, Castration-Resistant/drug therapy , Retrospective Studies , Treatment Outcome
3.
Eur J Cancer ; 87: 140-146, 2017 12.
Article in English | MEDLINE | ID: mdl-29149760

ABSTRACT

BACKGROUND: The GETUG 13 phase III trial tested personalised chemotherapy based on tumour marker decline in patients with poor-prognosis germ-cell tumour (GCT) and demonstrated that a dose-dense regimen improves progression-free survival in patients with an unfavourable decline. We investigated the pattern of relapse for patients included in GETUG 13. METHODS: We conducted an analysis of relapse events in patients from GETUG 13. Baseline procedures before inclusion in the trial comprised a thoraco-abdomino-pelvic computed tomography scan and a magnetic resonance imaging of the brain. RESULTS: With a median follow-up of 4.1 years (0.3; 8.8 years), a progression event was observed in 109/254 patients (43%). First event consisted in a marker progression only in 47 patients (43%), a radiographic progression only in 35 patients (32%), a mix progression on both markers and imaging in 12 patients (11%) and death in 15 patients (14%). In patients with radiographic progression only, brain was the predominant site (n = 19/35, 54%). Among patients with unfavourable decline who experienced a radiographic progression (as first and subsequent progression event, n = 58), brain was a site of progression in 28 patients (48%): 12/30 (40%) in patients treated with cisplatin, bleomycin and etoposide and 16/28 (57%) in those treated with dose-dense chemotherapy. CONCLUSIONS: Brain metastases develop often, early and frequently as the only site of relapse in the course of poor-prognosis GCT. This raises the question of early detection and optimal treatment of brain metastases in these patients, e.g. by integrating a systematic brain MRI after 2-3 months of chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/secondary , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Germ Cell and Embryonal/secondary , Testicular Neoplasms/drug therapy , Testicular Neoplasms/pathology , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/mortality , Clinical Trials, Phase III as Topic , Disease Progression , Disease-Free Survival , France , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Multicenter Studies as Topic , Neoplasms, Germ Cell and Embryonal/diagnostic imaging , Neoplasms, Germ Cell and Embryonal/mortality , Proportional Hazards Models , Retrospective Studies , Risk Factors , Testicular Neoplasms/diagnostic imaging , Testicular Neoplasms/mortality , Time Factors , Tomography, X-Ray Computed , Treatment Outcome , United States
5.
Oncogene ; 35(46): 5963-5976, 2016 11 17.
Article in English | MEDLINE | ID: mdl-26804168

ABSTRACT

Advanced prostate adenocarcinomas enriched in stem-cell features, as well as variant androgen receptor (AR)-negative neuroendocrine (NE)/small-cell prostate cancers are difficult to treat, and account for up to 30% of prostate cancer-related deaths every year. While existing therapies for prostate cancer such as androgen deprivation therapy (ADT), destroy the bulk of the AR-positive cells within the tumor, eradicating this population eventually leads to castration-resistance, owing to the continued survival of AR-/lo stem-like cells. In this study, we identified a critical nexus between p38MAPK signaling, and the transcription factor Forkhead Box Protein C2 (FOXC2) known to promote cancer stem-cells and metastasis. We demonstrate that prostate cancer cells that are insensitive to ADT, as well as high-grade/NE prostate tumors, are characterized by elevated FOXC2, and that targeting FOXC2 using a well-tolerated p38 inhibitor restores epithelial attributes and ADT-sensitivity, and reduces the shedding of circulating tumor cells in vivo with significant shrinkage in the tumor mass. This study thus specifies a tangible mechanism to target the AR-/lo population of prostate cancer cells with stem-cell properties.


Subject(s)
Drug Resistance, Neoplasm , Epithelium/metabolism , Epithelium/pathology , Forkhead Transcription Factors/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Androgens/metabolism , Animals , Benzamides , Cell Line, Tumor , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Forkhead Transcription Factors/genetics , Gene Expression , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Models, Biological , Neoplasm Grading , Nitriles , Phenotype , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Recurrence , Xenograft Model Antitumor Assays , Zinc Finger E-box-Binding Homeobox 1/metabolism
6.
Prostate Cancer Prostatic Dis ; 18(3): 276-80, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26171883

ABSTRACT

BACKGROUND: Prostate cancer persisting in the primary site after systemic therapy may contribute to emergence of resistance and progression. We previously demonstrated molecular characteristics of lethal cancer in the prostatectomy specimens of patients presenting with lymph node metastasis after chemohormonal treatment. Here we report the post-treatment outcomes of these patients and assess whether a link exists between surgery and treatment-free/cancer-free survival. METHODS: Patients with either clinically detected lymph node metastasis or primaries at high risk for nodal dissemination were treated with androgen ablation and docetaxel. Those responding with PSA concentration <1 ng ml(-1) were recommended surgery 1 year from enrollment. ADT was withheld postoperatively. The rate of survival without biochemical progression 1 year after surgery was measured to screen for efficacy. RESULTS: Forty patients were enrolled and 39 were evaluable. Three patients (7.7%) declined surgery. Of the remaining 36, 4 patients experienced disease progression during treatment and 4 more did not reach PSA <1. Twenty-six patients (67%) completed surgery, and 13 (33%) were also progression-free 1 year postoperatively (8 with undetectable PSA). With a median follow-up of 61 months, time to treatment failure was 27 months in the patients undergoing surgery. The most frequent patterns of first disease recurrence were biochemical (10 patients) and systemic (5). CONCLUSIONS: Half of the patients undergoing surgery were off treatment and progression-free 1 year following completion of all therapy. These results suggest that integration of surgery is feasible and may be superior to systemic therapy alone for selected prostate cancer patients presenting with nodal metastasis.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Prostatectomy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/surgery , Adult , Aged , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/adverse effects , Disease Progression , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local , Neoplasm Staging , Prostate-Specific Antigen , Prostatectomy/adverse effects , Prostatectomy/methods , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Treatment Failure , Treatment Outcome
7.
Ann Oncol ; 26(8): 1589-604, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26041764

ABSTRACT

The first St Gallen Advanced Prostate Cancer Consensus Conference (APCCC) Expert Panel identified and reviewed the available evidence for the ten most important areas of controversy in advanced prostate cancer (APC) management. The successful registration of several drugs for castration-resistant prostate cancer and the recent studies of chemo-hormonal therapy in men with castration-naïve prostate cancer have led to considerable uncertainty as to the best treatment choices, sequence of treatment options and appropriate patient selection. Management recommendations based on expert opinion, and not based on a critical review of the available evidence, are presented. The various recommendations carried differing degrees of support, as reflected in the wording of the article text and in the detailed voting results recorded in supplementary Material, available at Annals of Oncology online. Detailed decisions on treatment as always will involve consideration of disease extent and location, prior treatments, host factors, patient preferences as well as logistical and economic constraints. Inclusion of men with APC in clinical trials should be encouraged.


Subject(s)
Adenocarcinoma/therapy , Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Bone Density Conservation Agents/therapeutic use , Prostatic Neoplasms, Castration-Resistant/therapy , Prostatic Neoplasms/therapy , Taxoids/therapeutic use , Adenocarcinoma/pathology , Antineoplastic Agents/therapeutic use , Docetaxel , Humans , Male , Orchiectomy , Practice Guidelines as Topic , Prostatectomy , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/pathology , Radiotherapy, Adjuvant
8.
Br J Cancer ; 107(9): 1547-53, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23033003

ABSTRACT

BACKGROUND: The bone-forming metastases of prostate cancer result from complex stromal-epithelial interactions within the tumour microenvironment. Autocrine-paracrine signalling pathways between prostate cancer epithelial cells, osteoblasts, and osteoclasts stimulate aberrant bone remodelling, and the activity of these three cell populations can be quantitatively measured using prostate-specific antigen (PSA), bone-specific alkaline phosphatase (BAP) and urine N-telopeptide (uNTx), respectively. The purpose of the present study was to test the hypothesis that serial measurements of BAP and uNTx during therapy would facilitate monitoring of disease activity and predict the overall survival (OS) in patients with metastatic prostate cancer receiving therapy. METHODS: Radionuclide bone scan, PSA, BAP, and uNTx data were retrospectively analysed from three clinical trials in patients with metastatic prostate cancer conducted at our institution. Qualitative changes in bone scans and quantitative changes in PSA, BAP, and uNTx concentrations during therapy were correlated with OS. RESULTS: Baseline levels of BAP, but not PSA, were prognostic for OS in both androgen-dependent and castrate-resistant disease. A reduction in PSA, BAP, uNTx, or BAP/uNTx on therapy was predictive of improved OS in both patient groups. Conversely, an increase in PSA, or BAP on therapy was predictive of worse OS in both patient groups. Baseline number of lesions and response on bone scan during therapy were neither prognostic nor predictive of OS in either patient group. CONCLUSION: These observations support the concept that serial measurements of bone turnover metabolites during therapy function as clinically informative predictive biomarkers in patients with advanced prostate cancer and skeletal metastases. PSA measurements and bone scans remain essential to monitor the overall disease activity and determine the anatomic distribution of skeletal metastases.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/metabolism , Bone and Bones/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Alkaline Phosphatase/analysis , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Bone Remodeling , Bone and Bones/pathology , Collagen Type I/urine , Humans , Kallikreins/analysis , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Metastasis , Peptides/urine , Prognosis , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/drug therapy , Randomized Controlled Trials as Topic , Retrospective Studies , Survival Analysis
9.
Br J Cancer ; 107(4): 646-51, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22805323

ABSTRACT

BACKGROUND: Patients with prostate cancer tend to die from bone metastases. Until now, no evidence has shown that Paget's disease of bone (PDB) affects the progression of bone metastasis or overall survival of patients with prostate cancer. METHODS: We searched our patient database for men who had presented with prostate cancer and PDB between June 1993 and March 2009, and identified best-matched control patients according to stage, grade, age, date of diagnosis, treatment, and race. RESULTS: Among 1346 consecutive patients with prostate cancer diagnosed before 2008, 15 were confirmed to have comorbid PDB. Twenty-six more were identified from the institutional billing search. Including the 41 best-matched controls, our total study population was 82 patients. In the Kaplan-Meier analysis, we estimated median times from diagnosis of prostate cancer to bone metastasis to be 21.5 years for those with PDB and 9.4 years for those without PDB (P=0.044). Median overall survival times were 11.8 and 9.2 years for the two groups, respectively (P=0.008). CONCLUSION: For the first time, we have obtained evidence that patients with prostate cancer and PDB have delayed time to bone metastases and improved overall survival than do patients with prostate cancer alone.


Subject(s)
Bone Neoplasms/secondary , Osteitis Deformans/complications , Prostatic Neoplasms/complications , Aged , Case-Control Studies , Humans , Male , Middle Aged , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Time Factors
10.
Ann Oncol ; 23(1): 46-52, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21464158

ABSTRACT

BACKGROUND: We investigated cytokines and angiogenic factors (CAFs) in patients with metastatic renal cell carcinoma (mRCC) treated in a randomized phase II clinical trial of sorafenib versus sorafenib+ interferon-α (IFN-α) that yielded no differences in progression-free survival (PFS). We aimed to link the CAF profile to PFS and select candidate predictive and prognostic markers for further study. METHODS: The concentrations of 52 plasma CAFs were measured pretreatment (n = 69), day 28, and day 56 using multiplex bead arrays and enzyme-linked immunosorbent assay. We investigated the association between baseline levels of CAFs with PFS and posttreatment changes. RESULTS: Unsupervised CAF clustering analysis revealed two distinct mRCC patient groups with elevated proangiogenic or proinflammatory mediators. A six-marker baseline CAF signature [osteopontin, vascular endothelial growth factor (VEGF), carbonic anhydrase 9, collagen IV, VEGF receptor-2, and tumor necrosis factor-related apoptosis-inducing ligand] correlated with PFS benefit (hazard ratio 0.20 versus 2.25, signature negative versus positive, respectively; P = 0.0002). While changes in angiogenic factors were frequently attenuated by the sorafenib+ IFN combination, most key immunomodulatory mediators increased. CONCLUSIONS: Using CAF profiling, we identified two mRCC patient groups, a candidate plasma signature for predicting PFS benefit, and distinct marker changes occurring with each treatment. This platform may provide valuable insights into renal cell carcinoma biology and the molecular consequences of targeted therapies.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Renal Cell/blood , Cytokines/blood , Kidney Neoplasms/blood , Vascular Endothelial Growth Factor A/blood , Antineoplastic Agents/therapeutic use , Benzenesulfonates/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/mortality , Cluster Analysis , Disease-Free Survival , Enzyme-Linked Immunosorbent Assay , Humans , Interferon-alpha/administration & dosage , Kaplan-Meier Estimate , Kidney Neoplasms/drug therapy , Kidney Neoplasms/mortality , Niacinamide/analogs & derivatives , Phenylurea Compounds , Pyridines/therapeutic use , Sorafenib
11.
Ann Oncol ; 23(3): 688-694, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21821830

ABSTRACT

BACKGROUND: This phase 1/2 study assessed sunitinib combined with docetaxel (Taxotere) and prednisone in chemotherapy-naive metastatic, castration-resistant prostate cancer (mCRPC) patients. PATIENTS AND METHODS: To determine the recommended phase 2 dose (RP2D), 25 patients in four dose escalation cohorts received 3-week cycles of sunitinib (2 weeks on, 1 week off), docetaxel and prednisone, preceded by a 4-week sunitinib 50 mg/day lead in. RP2D was evaluated in 55 additional patients. The primary end point was prostate-specific antigen (PSA) response rate. RESULTS: One phase 1 dose-limiting toxicity occurred (grade 3 hyponatremia). The RP2D was sunitinib 37.5 mg/day, docetaxel 75 mg/m(2) and prednisone 5 mg b.i.d. During phase 2, confirmed PSA responses occurred in 31 patients [56.4% (95% confidence interval 42.3-69.7)]. Median time to PSA progression was 9.8 months. Forty-one patients (75%) were treated >3 months, 12 (22%) completed the study (16 cycles) and 43 (78%) discontinued (36% for disease progression and 27% adverse events). The most frequent treatment-related grade 3/4 adverse events were neutropenia (53%; 15% febrile) and fatigue/asthenia (16%). Among 33 assessable patients, 14 (42.4%) had confirmed partial response. Median progression-free and overall survivals were 12.6 and 21.7 months, respectively. CONCLUSION: This combination was moderately well tolerated, with promising response rate and survival benefit, justifying further investigation in mCRPC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Prostatic Neoplasms/drug therapy , Aged , Aged, 80 and over , Disease-Free Survival , Docetaxel , Drug Resistance, Neoplasm/drug effects , Humans , Indoles/administration & dosage , Indoles/adverse effects , Kaplan-Meier Estimate , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis/drug therapy , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Prednisone/administration & dosage , Prednisone/adverse effects , Prostate-Specific Antigen/blood , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Pyrroles/administration & dosage , Pyrroles/adverse effects , Sunitinib , Taxoids/administration & dosage , Taxoids/adverse effects
12.
Prostate Cancer Prostatic Dis ; 13(3): 252-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20386565

ABSTRACT

Prediction of cancer progression after radical prostatectomy is one of the most challenging problems in the management of prostate cancer. Gene-expression profiling is widely used to identify genes associated with such progression. Usually candidate genes are identified according to a gene-by-gene comparison of expression. Recent reports suggested that relative expression of a gene pair more efficiently predicts cancer progression than single-gene analysis does. The top-scoring pair (TSP) algorithm classifies phenotypes according to the relative expression of a pair of genes. We applied the TSP approach to predict, which patients would experience systemic tumor progression after radical prostatectomy. Relative expression of TPD52L2/SQLE and CEACAM1/BRCA1 gene pairs identified those patients with more than 99% specificity but relatively low sensitivity (approximately 10%). These two gene pairs were validated in three independent data sets. In addition, combining two pairs of genes improved sensitivity without compromising specificity. Functional annotation of the TSP genes showed that they cluster by a limited number of biological functions and pathways, suggesting that relatively lower expression of genes from specific pathways can predict cancer progression. In conclusion, comparative analysis of the expression of two genes may be a simple and effective classifier for prediction of prostate cancer progression. In summary, the TSP approach can be used to identify patients whose prostate cancer will progress after they undergo radical prostatectomy. Two gene pairs can predict which men would experience progression to the metastatic form of the disease. However, because our analysis was based on a relatively small number of genes, a larger study will be needed to identify the best predictors of disease outcome overall.


Subject(s)
Algorithms , Biomarkers, Tumor/genetics , Gene Expression Profiling , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Case-Control Studies , Computational Biology , Disease Progression , Humans , Male , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Prognosis , Sensitivity and Specificity , Survival Rate
13.
Prostate Cancer Prostatic Dis ; 13(1): 6-11, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19581923

ABSTRACT

Caveolin-1 (cav-1) is reportedly overexpressed in prostate cancer cells and is associated with disease progression. Specific oncogenic activities of cav-1 associated with Akt activation also occur in prostate cancer. A membrane-associated protein, cav-1, is nonetheless secreted by prostate cancer cells; results of recent studies showed that secreted cav-1 can stimulate cell survival and angiogenic activities, defining a role for cav-1 in the prostate cancer microenvironment. Serum cav-1 levels were also higher in prostate cancer patients than in control men without prostate cancer, and the preoperative serum cav-1 concentration had prognostic potential in men undergoing radical prostatectomy. Secreted cav-1 is therefore a potential biomarker and therapeutic target for prostate cancer.


Subject(s)
Caveolin 1/physiology , Prostatic Neoplasms/physiopathology , Biomarkers, Tumor , Caveolin 1/blood , Caveolin 1/metabolism , Disease Progression , Humans , Male , Neovascularization, Pathologic , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Up-Regulation
14.
Br J Cancer ; 99(9): 1426-32, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18841158

ABSTRACT

In a placebo-controlled randomised study of the platelet-derived growth factor receptor (PDGFR) inhibitor imatinib mesylate and docetaxel in metastatic prostate cancer with bone metastases (n=116), no significant differences in progression-free and overall survival were observed. To evaluate pharmacodynamic correlates of outcomes, we assessed the association of plasma platelet-derived growth factor (PDGF) isoform kinetics and PDGFR inhibition with progression-free and overall survival by individual treatment arm. We found that in the docetaxel-placebo arm alone, the probability of decrease in PDGFR phosphorylation (Pr-Decr-pPDGFR) above 0.5 (vs 30 months (HR 3.1; P=0.04 in log-rank test). By contrast, in the docetaxel plus imatinib arm, the association of Pr-Decr-pPDGFR >0.5 with a rise in plasma PDGF isoform concentrations and inferior survival was not observed. The data suggest that dynamic changes in PDGFR phosphorylation in peripheral blood leukocytes predict docetaxel efficacy. Rising plasma PDGF concentrations may explain and/or mark docetaxel resistance. Validation and mechanistic studies addressing these unexpected findings should anticipate a confounding influence of concurrent PDGFR inhibitor therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukocytes/metabolism , Prostatic Neoplasms/drug therapy , Receptors, Platelet-Derived Growth Factor/metabolism , Taxoids/therapeutic use , Dimerization , Docetaxel , Humans , Male , Multivariate Analysis , Phosphorylation , Platelet-Derived Growth Factor/analysis , Platelet-Derived Growth Factor/chemistry , Platelet-Derived Growth Factor/physiology , Prostatic Neoplasms/blood , Prostatic Neoplasms/mortality
15.
Oncogene ; 27(39): 5195-203, 2008 Sep 04.
Article in English | MEDLINE | ID: mdl-18490922

ABSTRACT

ErbB3 is a transmembrane growth factor receptor that has been implicated in the pathogenesis of human cancer. After finding that a truncated form of ErbB3 was present and upregulated in metastatic prostate cancer cells in lymph nodes and bone, we explored the pathophysiological functions of this unusual form of ErbB3 in the context of mouse calvaria as well as osteoblasts in vitro and the femur microenvironment in vivo. Here we demonstrate that prostate cancer cells expressed an alternatively spliced transcript that encodes a 45-kDa glycosylated protein (p45-sErbB3). The recombinant p45-sErbB3 purified from conditioned medium stimulated calvarial bone formation and induced osteoblast differentiation. Overexpression of p45-sErbB3 in the osteolytic prostate cancer cell line PC-3 converted its phenotype from bone lysing to bone forming upon injection into the femurs of immunodeficient mice. Further, we detected sErbB3 in plasma samples from patients with castration-resistant prostate cancer with bone metastasis. These observations establish that p45-sErbB3 is a structurally and functionally unique gene product of ErbB3 and suggest that p45-sErbB3 is likely one of the factors involved in the osteoblastic bone metastases of prostate cancer.


Subject(s)
Bone Development/physiology , Prostatic Neoplasms/metabolism , Receptor, ErbB-3/physiology , Alternative Splicing , Animals , Base Sequence , Bone Neoplasms/secondary , Cell Line, Tumor , Culture Media, Conditioned , DNA Primers , Humans , Male , Mice , Osteoblasts/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Oncogene ; 27(5): 596-603, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-17700537

ABSTRACT

The tendency of prostate cancer to produce osteoblastic bone metastases suggests that cancer cells and osteoblasts interact in ways that contribute to cancer progression. To identify factors that mediate these interactions, we compared gene expression patterns between two bone-derived prostate cancer cell lines that produce osteoblastic (MDA PCa 2b) or osteolytic lesions (PC-3). Both cell lines expressed Wnt ligands, including WNT7b, a canonical Wnt implicated in osteogenesis. PC-3 cells expressed 50 times more Dickkopf-1 (DKK1), an inhibitor of Wnt pathways, than did MDA PCa 2b cells. Evaluation of the functional role of these factors (in cocultures of prostate cancer cells with primary mouse osteoblasts (PMOs) or in bone organ cultures) showed that MDA PCa 2b cells activated Wnt canonical signaling in PMOs and that DKK1 blocked osteoblast proliferation and new bone formation induced by MDA PCa 2b cells. MDA PCa 2b cells did not induce bone formation in calvaria from mice lacking the Wnt co-receptor Lrp5. In human specimens, WNT7b was not expressed in normal prostate but was expressed in areas of high-grade prostate intraepithelial neoplasia, in three of nine primary prostate tumor specimens and in 16 of 38 samples of bone metastases from prostate cancer. DKK1 was not expressed in normal or cancerous tissue but was expressed in two of three specimens of osteolytic bone metastases (P=0.0119). We conclude that MDA PCa 2b induces new bone formation through Wnt canonical signaling, that LRP5 mediates this effect, and that DKK1 is involved in the balance between bone formation and resorption that determines lesion phenotype.


Subject(s)
Bone Neoplasms/secondary , LDL-Receptor Related Proteins/metabolism , Prostatic Neoplasms/metabolism , Wnt Proteins/metabolism , Animals , Bone Neoplasms/metabolism , Cell Line, Tumor , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5 , Male , Mice , Osteoblasts/metabolism , Osteolysis , Phenotype , Prostatic Neoplasms/pathology , Signal Transduction
17.
Oncogene ; 25(44): 6015-25, 2006 Sep 28.
Article in English | MEDLINE | ID: mdl-16682954

ABSTRACT

An inverse relationship exists between the expression of 15-lipoxygenase-2 (15-LOX-2) and peroxisome proliferator-activated receptor gamma (PPARgamma) in normal prostate epithelial cells (PrECs) compared with their expression in prostate carcinoma cells (PC-3). The reason for this difference, however, is unknown. We hypothesized that this inverse expression partly involves the 15-LOX-2 promoter and 15-S-hydroxyeicosatetraenoic acid (15-(S)-HETE), a product of 15-LOX-2 that binds to PPARgamma. We identified an active steroid nuclear receptor half-site present in the 15-LOX-2 promoter fragment F-5 (-618/+177) that can interact with PPARgamma. After forced expression of wild-type PPARgamma, 15-(S)-HETE (1 microM) decreased F-5 reporter activity in PrECs whereas forced expression of 15-LOX-2 resulted in 15-(S)-HETE production which enhanced F-5 activity in PC-3. In contrast, the expression of dominant-negative PPARgamma reversed the transcriptional activation of F-5 by enhancing it 202-fold in PrEC or suppressing it in PC-3; the effect in PC-3 was positively increased 150-fold in the presence of 15-(S)-HETE (1 microM). Peroxisome proliferator-activated receptor gamma interacted with 15-LOX-2 promoter sequences in pulldown experiments using biotinylated 15-LOX-2 (-560/-596 bp) oligonucleotides. In gelshift analyses PPARgamma and orphan receptor RORalpha were shown to interact with the F-5 fragment in PC-3 cells. These data suggest that crosstalk mechanisms exist between the 15-LOX-2 gene and PPARgamma to counterbalance expression and help explain the inverse relationship of these genes in normal versus cancer cells.


Subject(s)
Arachidonate 15-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/genetics , Down-Regulation/genetics , Feedback, Physiological/genetics , Hydroxyeicosatetraenoic Acids/physiology , PPAR gamma/physiology , 5' Untranslated Regions , Cell Line , Cell Line, Tumor , Chromosomes, Human, Pair 17/enzymology , Chromosomes, Human, Pair 17/genetics , Cloning, Molecular , Enhancer Elements, Genetic , Humans , Lipoxygenase Inhibitors , Male , Promoter Regions, Genetic , Prostate/cytology , Prostate/enzymology , Prostate/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor Cross-Talk/physiology , Up-Regulation/genetics
18.
Onkologie ; 27(4): 345-50, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15347888

ABSTRACT

INTRODUCTION: This retrospective study addressed the possible involvement of latent Epstein-Barr virus (EBV) infection, in particular LMP-1 expression, and further exogenous factors, i.e. tobacco, alcohol and occupational hazardous substances, in nasopharyngeal carcinoma (NPC) in a German population. PATIENTS AND METHODS: From 1980 to 2000, 44 patients suffering from histologically confirmed NPC were entered into the study. 33 specimens were available for immunostaining (IHC) to analyze LMP-1 expression. Information about environmental exposures were obtained employing a detailed standardized questionnaire. RESULTS: Outcome of patients with squamous cell NPC (SC-NPC) was significant worse than that of those with non-keratinizing NPC (NK-NPC). Age and tumor size correlated with response to therapy. The group with negative conventional LMP-1 staining showed better overall survival after 5 years compared to the group with positive or marginally positive LMP-1 detection (not significant). Nevertheless, after staining by tyramid-augmented IHC (TSA-IHC), nearly all specimens with negative LMP-1-staining in conventional IHC were found to be clearly positive. All patients with SC-NPC were smokers. The distribution of smokers and non-smokers in the group of NK-NPC was balanced. Comparable to the tobacco observation, there was also a correlation between high alcohol consumption and SC-NPC. CONCLUSION: Prognosis of NPC is mainly dependent on histologic type. Prognostic impact of LMP-1 is still unclear since LMP-1 was detected in all specimens using TSA-IHC. Therefore, TSA-IHC-LMP-1 detection might be interesting for diagnostic specification and development of new therapeutic strategies in NPC.


Subject(s)
Carcinoma, Squamous Cell/diagnosis , Carrier Proteins/analysis , Epstein-Barr Virus Infections/diagnosis , Nasopharyngeal Neoplasms/diagnosis , Slow Virus Diseases/diagnosis , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Alcohol Drinking/adverse effects , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/pathology , Cocarcinogenesis , Cross-Sectional Studies , Cytoskeletal Proteins , Epstein-Barr Virus Infections/epidemiology , Epstein-Barr Virus Infections/pathology , Female , Germany , Humans , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Male , Middle Aged , Nasopharyngeal Neoplasms/epidemiology , Nasopharyngeal Neoplasms/pathology , Neoplasm Staging , Prognosis , Retrospective Studies , Risk Factors , Slow Virus Diseases/epidemiology , Slow Virus Diseases/pathology , Smoking/adverse effects
19.
Br J Cancer ; 86(10): 1555-60, 2002 May 20.
Article in English | MEDLINE | ID: mdl-12085204

ABSTRACT

Only about half of patients with a poor-prognosis non-seminomatous germ-cell tumours can achieve a cure. The aim of this phase II study was to assess the efficacy and toxicity of a dose-dense alternating chemotherapy regimen in this subset of patients. High volume non-seminomatous germ-cell tumours was defined as follows: at least two sites of non pulmonary metastases, an extragonadal primary tumour, a serum human chorionic gonadotropin level higher than 10 000 mIU x ml(-1), or a alpha-foetoprotein level higher than 2000 mIU ml(-1). Patients who fulfilled these criteria were treated with the so-called BOP-CISCA-POMB-ACE regimen (bleomycin, vincristine, and cisplatin; cisplatin, cyclophosphamide, and doxorubicin; cisplatin, vincristine, methotrexate, and bleomycin; etoposide, dactinomycin, and cyclophosphamide) plus granulocyte colony-stimulating factor. A total of 58 patients were enrolled. Patients were retrospectively classified according to the International Germ-Cell Cancer Consensus Group classification; 38 patients (66%) had poor-prognosis disease and 19 patients (33%) had intermediate-prognosis. Patients received a median of 2.5 courses (range 0.25 to five courses) of the BOP-CISCA-POMB-ACE regimen. Forty-two patients (72.4%) had a complete response to therapy. With a median follow-up time of 31 months, the 3-year progression-free survival rate was 71% (95% confidence interval, 60 to 84%) and the 3-year overall survival rate was 73% (95% confidence interval: 62 to 86%). The 3-year PFS rates were 83% (95% confidence interval: 68 to 100%) in the intermediate-prognosis group and 65% (95% confidence interval: 51 to 82%) in the poor-prognosis group. Early side effects included mainly grade 4 haematologic toxicity (neutropaenia in 79% of patients, thrombocytopaenia in 69%, anaemia in 22%), grade 4 stomatitis (19%), and four early deaths (7% of patients), at least partially related to toxicity. The dose-dense BOP-CISCA-POMB-ACE regimen is highly active in patients with non-seminomatous germ-cell tumours classified as intermediate-prognosis or poor-prognosis according to the International Germ-Cell Cancer Consensus Group. Because outcomes with this regimen compare favourably with outcome after standard therapy, dose-dense chemotherapy should be further investigated in this subset of patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Germinoma/drug therapy , Seminoma/drug therapy , Testicular Neoplasms/drug therapy , Adolescent , Adult , Anemia, Refractory, with Excess of Blasts/chemically induced , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor/blood , Bleomycin/administration & dosage , Bleomycin/adverse effects , Cisplatin/administration & dosage , Cisplatin/adverse effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Dactinomycin/administration & dosage , Dactinomycin/adverse effects , Disease-Free Survival , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Etoposide/administration & dosage , Etoposide/adverse effects , Gastrointestinal Diseases/chemically induced , Germinoma/mortality , Germinoma/pathology , Germinoma/secondary , Hematologic Diseases/chemically induced , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Male , Methotrexate/administration & dosage , Methotrexate/adverse effects , Middle Aged , Peripheral Nervous System Diseases/chemically induced , Prognosis , Prospective Studies , Remission Induction , Seminoma/mortality , Seminoma/pathology , Seminoma/secondary , Survival Analysis , Survival Rate , Testicular Neoplasms/mortality , Testicular Neoplasms/pathology , Treatment Outcome , Vincristine/administration & dosage , Vincristine/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...