Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 70(8): 3239-48, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20388787

ABSTRACT

To identify potential mechanisms underlying prostate cancer chemotherapy response and resistance, we compared the gene expression profiles in high-risk human prostate cancer specimens before and after neoadjuvant chemotherapy and radical prostatectomy. Among the molecular signatures associated with chemotherapy, transcripts encoding inhibitor of DNA binding 1 (ID1) were significantly upregulated. The patient biochemical relapse status was monitored in a long-term follow-up. Patients with ID1 upregulation were found to be associated with longer relapse-free survival than patients without ID1 increase. This in vivo clinical association was mechanistically investigated. The chemotherapy-induced ID1 upregulation was recapitulated in the prostate cancer cell line LNCaP. Docetaxel dose-dependently induced ID1 transcription, which was mediated by ID1 promoter E-box chromatin modification and c-Myc binding. Stable ID1 overexpression in LNCaP increased cell proliferation, promoted G(1) cell cycle progression, and enhanced docetaxel-induced cytotoxicity. These changes were accompanied by a decrease in cellular mitochondria content, an increase in BCL2 phosphorylation at serine 70, caspase-3 activation, and poly(ADP-ribose) polymerase cleavage. In contrast, ID1 siRNA in the LNCaP and C42B cell lines reduced cell proliferation and decreased docetaxel-induced cytotoxicity by inhibiting cell death. ID1-mediated chemosensitivity enhancement was in part due to ID1 suppression of p21. Overexpression of p21 in LNCaP-ID1-overexpressing cells restored the p21 level and reversed ID1-enhanced chemosensitivity. These molecular data provide a mechanistic rationale for the observed in vivo clinical association between ID1 upregulation and relapse-free survival. Taken together, it shows that ID1 expression has a novel therapeutic role in prostate cancer chemotherapy and prognosis.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation, Neoplastic , Inhibitor of Differentiation Protein 1/metabolism , Prostatic Neoplasms/drug therapy , Taxoids/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Chromatin/chemistry , Docetaxel , Dose-Response Relationship, Drug , G1 Phase , Humans , Male , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-myc/metabolism
2.
Prostate ; 70(4): 433-42, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19866475

ABSTRACT

BACKGROUND: Metastatic prostate cancer is either inherently resistant to chemotherapy or rapidly acquires this phenotype after chemotherapy exposure. In this study, we identified a docetaxel-induced resistance mechanism centered on CCL2. METHODS: We compared the gene expression profiles in individual human prostate cancer specimens before and after exposure to chemotherapy collected from previously untreated patients who participated in a clinical trial of preoperative chemotherapy. Subsequently, we used the gain- and loss-of-function approach in vitro to identify a potential mechanism underlying chemotherapy resistance. RESULTS: Among the molecular signatures associated with treatment, several genes that regulate the inflammatory response and chemokine activity were upregulated including a significant increase in transcripts encoding the CC chemokine CCL2. Docetaxel increased CCL2 expression in prostate cancer cell lines in vitro. CCL2-specific siRNA inhibited LNCaP and LAPC4 cell proliferation and enhanced the growth inhibitory effect of low-dose docetaxel. In contrast, overexpression of CCL2 or recombinant CCL2 protein stimulated prostate cancer cell proliferation and rescued cells from docetaxel-induced cytotoxicity. This protective effect of CCL2 was associated with activation of the ERK/MAP kinase and PI3K/AKT, inhibition of docetaxel-induced Bcl2 phosphorylation at serine 70, phosphorylation of Bad, and activation of caspase-3. The addition of a PI3K/AKT inhibitor Ly294002 reversed the CCL2 protection and was additive to docetaxel-induced toxicity. CONCLUSION: These results support a mechanism of chemotherapy resistance mediated by cellular stress responses involving the induction of CCL2 expression and suggest that inhibiting CCL2 activity could enhance therapeutic responses to taxane-based therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Chemokine CCL2/genetics , Gene Expression/drug effects , Prostatic Neoplasms/drug therapy , Taxoids/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/pharmacology , Docetaxel , Drug Resistance, Neoplasm/genetics , Drug Therapy, Combination , Gene Expression Profiling , Gene Silencing , Humans , Male , Mitoxantrone/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/pharmacology , Up-Regulation
3.
Clin Cancer Res ; 14(11): 3562-70, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18519790

ABSTRACT

1,25-Dihydroxyvitamin D3 [1,25(OH)2D3], the most active metabolite of vitamin D3, has significant antitumor activity in a broad range of preclinical models of cancer. In this study, we show that the Iroquois homeobox gene 5 (Irx5) is down-regulated by 1,25(OH)2D3 in human prostate cancer samples from patients randomly assigned to receive weekly high-dose 1,25(OH)2D3 or placebo before radical prostatectomy. Down-regulation of Irx5 by 1,25(OH)2D3 was also shown in the human androgen-sensitive prostate cancer cell line LNCaP and in estrogen-sensitive MCF-7 breast cancer cells. Knockdown of Irx5 by RNA interference showed a significant reduction in LNCaP cell viability, which was accompanied by an increase in p21 protein expression, G2-M arrest, and an increase in apoptosis. The induced apoptosis was partially mediated by p53, and p53 protein expression was increased as a result of Irx5 knockdown. Cell survival was similarly reduced by Irx5 knockdown in the colon cancer cell line HCT 116 and in MCF-7 breast cancer cells, each being derived from clinical tumor types that seem to be inhibited by 1,25(OH)2D3. Overexpression of Irx5 led to a reduction of p21 and p53 expression. This is the first report that Irx5 is regulated by 1,25(OH)2D3 in humans and the first report to show that Irx5 is involved in the regulation of both the cell cycle and apoptosis in human prostate cancer cells. Irx5 may be a promising new therapeutic target in cancer treatment.


Subject(s)
Apoptosis/genetics , Calcitriol/pharmacology , Cell Cycle/genetics , Homeodomain Proteins/drug effects , Prostatic Neoplasms/drug therapy , Transcription Factors/drug effects , Vitamins/pharmacology , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Down-Regulation , Female , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Prostatic Neoplasms/metabolism , RNA Interference , Randomized Controlled Trials as Topic , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/drug effects
4.
Anticancer Res ; 26(4A): 2647-51, 2006.
Article in English | MEDLINE | ID: mdl-16886675

ABSTRACT

Calcitriol, the principal active metabolite of vitamin D and a naturally occurring hormone, showed significant antineoplastic activity in pre-clinical models of prostate cancer and many other tumor types. These antineoplastic effects were observed at calcitriol concentrations substantially above the physiological range. While a number of mechanisms of action have been postulated, the induction of apoptosis and inhibition of proliferation have been most extensively reported. These pre-clinical findings motivated several investigators to pursue a series of clinical trials to examine the potential of targeting the vitamin D receptor for cancer treatment using calcitriol. Initial studies tested daily dosing of calcitriol and showed that substantial dose escalation was not feasible due to hypercalciuria and/or hypercalcemia. In contrast, weekly dosing of calcitriol allowed substantial dose escalation without dose-limiting toxicities. Notably, however, the commercially available formulation of calcitriol exhibited nonlinear pharmacokinetics at the highest doses tested. While substantially higher concentrations were achieved, the maximum tolerated dose was not established due to this pharmacological limitation. Intermittently-dosed calcitriol was then combined with several antineoplastic agents, including steroids, bisphosphonates and chemotherapeutic agents. The activity seen in a phase II study of weekly calcitriol plus docetaxel was particularly encouraging and led to the development of DN-101, a proprietary formulation designed for cancer treatment. DN-101 in combination with docetaxel is being evaluated in a placebo-controlled randomized clinical trial that has completed accrual.


Subject(s)
Calcitriol/therapeutic use , Prostatic Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Calcitriol/administration & dosage , Clinical Trials as Topic , Drug Administration Schedule , Humans , Male , Randomized Controlled Trials as Topic
5.
Clin Cancer Res ; 12(9): 2812-6, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16675575

ABSTRACT

BACKGROUND: Calcitriol, the natural ligand for the vitamin D receptor, has significant potential in prostate cancer treatment. Measurement of its antineoplastic activity in prostate cancer clinical trials may be complicated by effects of calcitriol on prostate-specific antigen (PSA) production. We examined the effects of calcitriol at similar concentration on cell proliferation, androgen receptor (AR) expression, and PSA production in vitro and on PSA concentrations in prostate cancer patients. EXPERIMENTAL DESIGN: LNCaP prostate cancer cell proliferation was examined by cell counts 6 days after exposure to a range of concentrations of calcitriol. AR and PSA protein was quantified in LNCaP cells over 96 hours after exposure to 1 nmol/L calcitriol. Serum PSA and free PSA was serially measured by immunoassay over a period of 8 days in patients with hormone-naïve prostate cancer after a single dose of 0.5 microg/kg calcitriol. RESULTS: Calcitriol treatment resulted in dose-dependent growth inhibition of LNCaP with approximately 50% growth inhibition at the clinically achievable concentration of 1 nmol/L. Time-dependent up-regulation of AR expression and of PSA production in LNCaP cells was shown at the same concentration. No significant change in serum PSA or free PSA over 8 days was seen in eight subjects treated with a single dose of 0.5 microg/kg calcitriol. The analysis was powered to detect a 1.23-fold change between the baseline and day 8 serum PSA. CONCLUSIONS: At clinically achievable concentrations, calcitriol inhibits growth and induces AR and PSA expression in LNCaP cells. We did not detect similar changes in serum PSA or free PSA in patients exposed to similar concentrations of calcitriol. Thus, a PSA flare, predicted by preclinical systems, is unlikely to occur in patients and therefore unlikely to complicate interpretation of clinical trial outcomes.


Subject(s)
Calcitriol/pharmacology , Prostate-Specific Antigen/metabolism , Cell Division/drug effects , Cell Line, Tumor , Humans , Kinetics , Male , Prostate-Specific Antigen/blood , Prostate-Specific Antigen/drug effects , Prostatic Neoplasms
6.
Urol Oncol ; 24(3): 254-9, 2006.
Article in English | MEDLINE | ID: mdl-16678060

ABSTRACT

PURPOSE: Currently available treatment modalities for high-risk clinically localized prostate cancer have limited chances of achieving complete tumor elimination because of either inadequate local or metastatic tumor eradication. The goal of this phase I/II study is to evaluate the safety and efficacy of neoadjuvant docetaxel and mitoxantrone before prostatectomy. MATERIALS AND METHODS: A total of 22 men with high-risk clinically localized prostate cancer underwent weekly treatment with docetaxel (35 mg/m(2)), with increasing doses of mitoxantrone (2-5 mg/m(2)) for a 12 of 16-week treatment cycle before prostatectomy. Testosterone and prostate-specific antigen (PSA) measurements were made before and after chemotherapy. RESULTS: The maximally tolerated dose for mitoxantrone was 4 mg/m(2), and the primary toxicity was neutropenia. Testosterone levels were maintained throughout treatment. PSA reductions were observed in 95% of patients, with a median reduction of 41%. The surgery was well tolerated after chemotherapy, without any major complications. Negative surgical margins were attained in 76% of patients. CONCLUSIONS: Administration of multi-agent chemotherapy before prostatectomy was safe in this population. This regimen appeared to have antineoplastic activity as evidenced by PSA reductions in the absence of significant testosterone changes. The benefit of chemotherapy for improving surgical margin rates could not be determined outside of a phase III trial because the effect of patient or surgeon factors could not be dissected from the potential effect of neoadjuvant therapy. Continued study of novel agents in the neoadjuvant setting is warranted because this approach allows for the rapid identification of active agents and for molecular investigation into the mechanism of drug activity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms/drug therapy , Aged , Docetaxel , Humans , Male , Maximum Tolerated Dose , Middle Aged , Mitoxantrone/administration & dosage , Neoadjuvant Therapy , Prostate-Specific Antigen/metabolism , Prostatectomy , Prostatic Neoplasms/surgery , Risk Factors , Taxoids/administration & dosage , Treatment Outcome
7.
World J Urol ; 23(1): 28-32, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15668801

ABSTRACT

Calcitriol, the principal active metabolite of vitamin D, has significant antineoplastic activity in pre-clinical models of prostate cancer and many other tumor types. Reported mechanisms of activity include inhibition of proliferation and cell cycle arrest, induction of apoptosis, and reduction of invasiveness and angiogenesis. Different mechanisms may be responsible in different tumor types and under different experimental conditions. Importantly, preclinical data suggest that calcitriol acts in a synergistic and/or additive manner when combined with antineoplastic agents that are relevant to prostate cancer, including dexamethasone and several classes of cytotoxic agents. The antineoplastic effects of calcitriol occur at concentrations that substantially exceed the normal physiologic range and cannot be safely achieved with conventional daily dosing. Intermittent administration of calcitriol has allowed significant dose escalation. In combination with weekly docetaxel, the agent produced encouraging results in a single-institution phase II study. An international placebo-controlled randomized trial that is currently under way will provide more robust information about the safety and efficacy of this combination.


Subject(s)
Androgens/metabolism , Calcitriol/therapeutic use , Calcium Channel Agonists/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Treatment Outcome
8.
Cancer Epidemiol Biomarkers Prev ; 13(12): 2225-32, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15598784

ABSTRACT

BACKGROUND: Cancer chemoprevention trials require enormous resources due to the large numbers of patients and the years of follow-up needed to achieve sufficient statistical power. Examination of candidate prevention agents using biomarkers as surrogate end points has been proposed as a method to rapidly identify promising agents for prevention trials. Treatment of patients with candidate agents prior to scheduled biopsy or surgical resection of malignancy allows for direct examination of the treatment effects on tumor tissue. In this study, we selected this approach to test several hypotheses about the effect of calcitriol (1,25-dihydroxycholecalciferol), the active form of vitamin D, on early-stage human prostate cancer. METHODS: After selection of surgical treatment for histologically confirmed adenocarcinoma of the prostate, patients were randomized to either calcitriol 0.5 mug/kg or placebo weekly for 4 weeks. The expression levels of the vitamin D receptor (VDR), proliferating cell nuclear antigen, PTEN (MMAC1/TEP1), c-Myc, transforming growth factor (TGF) beta receptor type II (TGFbeta RII), and Bcl-2 were quantified using immunohistochemistry in the patients' prostate specimens post surgery. RESULTS: Thirty-seven of 39 prostate tumors were evaluable for molecular end points. VDR expression was reduced in patients treated with calcitriol (mean, 75.3% of cells) compared with those that received placebo (mean, 98.6%; P = 0.005). Calcitriol treatment did not result in a statistically significant change in the fraction of cells expressing TGFbeta RII, PTEN, or proliferating cell nuclear antigen. Bcl-2 and c-Myc expression was at the lower limits of detection in both the calcitriol group and the placebo group; therefore, we were unable to determine whether drug treatment induced a significant change in these biomarkers. CONCLUSIONS: High-dose calcitriol down-regulates VDR expression in human prostate cancer. Further study is needed to determine the biological consequences of VDR down-regulation in prostate cancer. This study shows that the use of the preprostatectomy model is feasible and can be used to test the effect of candidate chemopreventive agents on prostate cancer.


Subject(s)
Adenocarcinoma/pathology , Adenocarcinoma/surgery , Biomarkers, Tumor/analysis , Calcitriol/pharmacology , Calcium Channel Agonists/pharmacology , Prostatectomy , Prostatic Neoplasms/surgery , Aged , Calcitriol/administration & dosage , Calcium Channel Agonists/administration & dosage , Humans , Immunohistochemistry , Male , Middle Aged , Placebos , Preoperative Care , Prostatic Neoplasms/pathology , Receptors, Calcitriol/biosynthesis
9.
Mol Cancer Ther ; 3(3): 373-81, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15026558

ABSTRACT

1,25-Dihydroxyvitamin D (calcitriol), the most active metabolite of vitamin D, has significant antineoplastic activity in preclinical models. Several mechanisms of activity have been proposed. These include inhibition of proliferation associated with cell cycle arrest and, in some models, differentiation, reduction in invasiveness and angiogenesis, and induction of apoptosis. Proposed mechanisms differ between tumor models and experimental conditions, and no unifying hypothesis about the mechanism of antineoplastic activity has emerged. Synergistic and/or additive effects with cytotoxic chemotherapy, radiation, and other cancer drugs have been reported. Significantly supraphysiological concentrations of calcitriol are required for antineoplastic effects. Such concentrations are not achievable in patients when calcitriol is dosed daily due to predictable hypercalcemia and hypercalcuria; however, phase I trials have demonstrated that intermittent dosing allows substantial dose escalation and has produced potentially therapeutic peak calcitriol concentrations. Recently, a phase II study reported encouraging levels of activity for the combination of high-dose calcitriol and docetaxel administered on a weekly schedule in patients with androgen-independent prostate cancer. This regimen is now under study in a placebo-controlled randomized trial in androgen-independent prostate cancer and in phase II studies in several other tumor types. Further work is needed to elucidate the molecular mechanisms of antineoplastic activity and optimal clinical applications of calcitriol in cancer.


Subject(s)
Calcitriol/pharmacology , Neoplasms/drug therapy , Animals , Apoptosis , Calcitriol/analogs & derivatives , Calcium Channel Agonists/pharmacology , Cell Differentiation , Cell Division , Clinical Trials as Topic , Humans , Ligands , Models, Biological , Neoplasms/metabolism , Neovascularization, Pathologic , Random Allocation , Signal Transduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...