Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Ann Oncol ; 31(8): 991-1000, 2020 08.
Article in English | MEDLINE | ID: mdl-32348852

ABSTRACT

BACKGROUND: Kinase fusions are rare and poorly characterized in breast cancer (BC). We aimed to characterize kinase fusions within a large cohort of advanced BC. PATIENTS AND METHODS: A total of 4854 patients with BC were analyzed by Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) targeted DNAseq and MSK-Fusion targeted RNAseq during the study time period. RESULTS: Twenty-seven of 4854 (0.6%) patients harbored fusions: 11 FGFR (five FGFR2, three FGFR3, three FGFR1), five BRAF, four NTRK1, two RET, two ROS1, one ALK, one ERBB2, and one MET. A history of endocrine therapy was present in 15 (56%) of fusion-positive BC; eight of the 15 cases had available pre-treatment samples, of which six were fusion-negative. None of the fusion-positive BC samples harbored ESR1 hotspot mutations. Two patients with acquired LMNA-NTRK1 fusions and metastatic disease received larotrectinib and demonstrated clinical benefit. CONCLUSION: Kinase fusions in BC are extremely rare, and appear to be enriched in hormone-resistant, metastatic carcinomas and mutually exclusive with ESR1 mutations. The present study expands the spectrum of genetic alterations activating mitogen-activated protein kinase (MAPK) signaling that can substitute for ESR1 mutations in this setting. Molecular testing at progression after endocrine therapy should include fusion testing, particularly in the absence of ESR1 hotspot alterations, in an effort to identify additional therapeutic options which may provide substantial clinical benefit.


Subject(s)
Breast Neoplasms , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Humans , Mutation , Proto-Oncogene Proteins
2.
Ann Oncol ; 30(Suppl_10): x3-x11, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31859348

ABSTRACT

One of the hallmarks of hormone receptor (HR)-positive breast cancer is its dependence on the phosphatidylinositol-3-kinase (PI3K) pathway. Here, we review the epidemiologic, functional, and pharmacologic interactions between oncogenic PI3K and the estrogen receptor (ER). We discuss the epidemiology of PI3K pathway alterations, mechanisms of resistance to PI3K inhibitors, and the current mechanistic landscape of crosstalk between PI3K and ER, which provide the rationale for dual ER and PI3K inhibition and is now a standard of care in the treatment of ER+ PIK3CA-mutant metastatic breast cancer. We outline newer studies in this field that delineate the clinically relevant overlaps between PI3K and parallel signaling pathways, insulin signaling, and ER epigenetic modifiers. We also identify several caveats with the current data and propose new strategies to overcome these bottlenecks.


Subject(s)
Breast Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Estrogen/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Drug Resistance, Neoplasm , Estrogen Receptor Modulators/administration & dosage , Estrogen Receptor Modulators/pharmacology , Female , Humans , Molecular Epidemiology , Molecular Targeted Therapy , Phosphoinositide-3 Kinase Inhibitors/administration & dosage , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Signal Transduction/drug effects
3.
Ann Oncol ; 30(9): 1417-1427, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31268127

ABSTRACT

BACKGROUND: NTRK1, NTRK2 and NTRK3 fusions are present in a plethora of malignancies across different histologies. These fusions represent the most frequent mechanism of oncogenic activation of these receptor tyrosine kinases, and biomarkers for the use of TRK small molecule inhibitors. Given the varying frequency of NTRK1/2/3 fusions, crucial to the administration of NTRK inhibitors is the development of optimal approaches for the detection of human cancers harbouring activating NTRK1/2/3 fusion genes. MATERIALS AND METHODS: Experts from several Institutions were recruited by the European Society for Medical Oncology (ESMO) Translational Research and Precision Medicine Working Group (TR and PM WG) to review the available methods for the detection of NTRK gene fusions, their potential applications, and strategies for the implementation of a rational approach for the detection of NTRK1/2/3 fusion genes in human malignancies. A consensus on the most reasonable strategy to adopt when screening for NTRK fusions in oncologic patients was sought, and further reviewed and approved by the ESMO TR and PM WG and the ESMO leadership. RESULTS: The main techniques employed for NTRK fusion gene detection include immunohistochemistry, fluorescence in situ hybridization (FISH), RT-PCR, and both RNA-based and DNA-based next generation sequencing (NGS). Each technique has advantages and limitations, and the choice of assays for screening and final diagnosis should also take into account the resources and clinical context. CONCLUSION: In tumours where NTRK fusions are highly recurrent, FISH, RT-PCR or RNA-based sequencing panels can be used as confirmatory techniques, whereas in the scenario of testing an unselected population where NTRK1/2/3 fusions are uncommon, either front-line sequencing (preferentially RNA-sequencing) or screening by immunohistochemistry followed by sequencing of positive cases should be pursued.


Subject(s)
Membrane Glycoproteins/isolation & purification , Neoplasms/diagnosis , Oncogene Proteins, Fusion/isolation & purification , Receptor, trkA/isolation & purification , Receptor, trkB/isolation & purification , Receptor, trkC/isolation & purification , Biomarkers, Tumor/genetics , Biomarkers, Tumor/isolation & purification , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry/standards , In Situ Hybridization, Fluorescence/standards , Medical Oncology/standards , Membrane Glycoproteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Precision Medicine/standards , Protein Kinase Inhibitors/therapeutic use , Receptor, trkA/genetics , Receptor, trkB/genetics , Receptor, trkC/genetics , Translational Research, Biomedical/standards
4.
Oncogene ; 36(16): 2255-2264, 2017 04 20.
Article in English | MEDLINE | ID: mdl-27748766

ABSTRACT

Dysregulated activation of the CDK4/6 kinases is a hallmark of most mammary-derived carcinomas. ATP-competitive inhibitors against this complex have been recently advanced in the clinic and have shown significant activity, particularly against tumors driven by the estrogen receptor (ER). However, resistance to these compounds has begun to emerge often months to years after their initiation. We investigated potential mechanisms of resistance using cell line models that are highly sensitive to this class of drugs. After prolonged exposure to the selective and potent CDK4/6 inhibitor LY2835219, clones emerged and several were found to harbor amplification of the CDK6 kinase. Amplification of CDK6 resulted in a marked increase in CDK6 expression and reduced response of the CDK4/6 target, phospho-Rb (pRb), to CDK4/6 inhibitors. Knockdown of CDK6 restored drug sensitivity, while enforced overexpression of CDK6 was sufficient to mediate drug resistance. Not only did CDK6 overexpression mediate resistance to CDK4/6 inhibitors but it also led to reduced expression of the ER and progesterone receptor (PR), and diminished responsiveness to ER antagonism. The reduced ER/PR expression after CDK4/6 inhibitor resistance was additionally observed in tumor biopsy specimens from patients treated with these drugs. Alternative mechanisms of resistance to CDK4/6 inhibitors such as loss of pRb and cyclin E1 overexpression also exhibited decreased hormone responsiveness, suggesting that the clinical paradigm of sequential endocrine-based therapy may be ineffective in some settings of acquired CDK4/6 resistance.


Subject(s)
Breast Neoplasms/enzymology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Resistance, Neoplasm , Protein Kinase Inhibitors/pharmacology , Receptors, Estrogen/metabolism , Aminopyridines/pharmacology , Benzimidazoles/pharmacology , Breast Neoplasms/diet therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 6/genetics , Female , Gene Amplification , Humans , MCF-7 Cells , Signal Transduction/drug effects
5.
Ann Oncol ; 26(7): 1494-500, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25851628

ABSTRACT

BACKGROUND: Assessment of phosphatase and tensin homolog deleted from chromosome 10 (PTEN) might be an important tool in identifying human epidermal growth factor receptor 2 (HER2)-positive breast cancer patients unlikely to derive benefit from anti-HER2 therapies. However, studies to date have failed to demonstrate its predictive role in any treatment setting. PATIENTS AND METHODS: Prospectively collected baseline core biopsies from 429 early-stage HER2-positive breast cancer patients treated with trastuzumab, lapatinib, or their combination in the Neo-ALTTO study were stained using two anti-PTEN monoclonal antibodies (CST and DAKO). The association of PTEN status and PI3K pathway activation (defined as either PTEN loss and/or PIK3CA mutation) with total pathological complete response (tpCR) at surgery, event-free survival (EFS), and overall survival (OS) was evaluated. RESULTS: PTEN loss was observed in 27% and 29% of patients (all arms, n = 361 and n = 363) for CST and DAKO, respectively. PTEN loss was more frequently observed in hormone receptor (HR)-negative (33% and 36% with CST and DAKO, respectively) compared with HR-positive tumours (20% and 22% with CST and DAKO, respectively). No significant differences in tpCR rates were observed according to PTEN status. PI3K pathway activation was found in 47% and 48% of patients (all arms, n = 302 and n = 301) for CST and DAKO, respectively. Similarly, tpCR rates were not significantly different for those with or without PI3K pathway activation. Neither PTEN status nor PI3K pathway activation were predictive of tpCR, EFS, or OS, independently of treatment arm or HR status. High inter-antibody and inter-observer agreements were found (>90%). Modification of scoring variables significantly affected the correlation between PTEN and HR status but not with tpCR. CONCLUSION: These data show that PTEN status determination is not a useful biomarker to predict resistance to trastuzumab and lapatinib-based therapies. The lack of standardization of PTEN status determination may influence correlations between expression and relevant clinical end points. CLINICAL TRIALS: This trial is registered with ClinicalTrials.gov: NCT00553358.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Chromosomes, Human, Pair 10/genetics , Gene Deletion , Neoadjuvant Therapy , PTEN Phosphohydrolase/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/pathology , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lapatinib , Neoplasm Staging , PTEN Phosphohydrolase/metabolism , Polymerase Chain Reaction , Prognosis , Prospective Studies , Quinazolines/administration & dosage , Remission Induction , Trastuzumab/administration & dosage
7.
Oncogene ; 33(26): 3422-31, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-23912457

ABSTRACT

Patients with HER2-positive breast cancer often exhibit intrinsic or acquired resistance to trastuzumab treatment. The transmembrane mucin 1 (MUC1) oncoprotein is aberrantly overexpressed in breast cancer cells and associates with HER2. The present studies demonstrate that silencing MUC1 C-terminal subunit (MUC1-C) in HER2-overexpressing SKBR3 and BT474 breast cancer cells results in the downregulation of constitutive HER2 activation. Moreover, treatment with the MUC1-C inhibitor, GO-203, was associated with disruption of MUC1-C/HER2 complexes and decreases in tyrosine-phosphorylated HER2 (p-HER2) levels. In studies of trastuzumab-resistant SKBR3R and BT474R cells, we found that the association between MUC1-C and HER2 is markedly increased (∼20-fold) as compared with that in sensitive cells. In addition, silencing MUC1-C in the trastuzumab-resistant cells or treatment with GO-203 decreased p-HER2 and AKT activation. Moreover, targeting MUC1-C was associated with the downregulation of phospho-p27 and cyclin E, which confer trastuzumab resistance. Consistent with these results, targeting MUC1-C inhibited the growth and clonogenic survival of both trastuzumab-resistant cells. Our results further demonstrate that silencing MUC1-C reverses resistance to trastuzumab and that the combination of GO-203 and trastuzumab is highly synergistic. These findings indicate that MUC1-C contributes to constitutive activation of the HER2 pathway and that targeting MUC1-C represents a potential approach to abrogate trastuzumab resistance.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Mucin-1/genetics , Receptor, ErbB-2/metabolism , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin E/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Lapatinib , Mice , Mice, Inbred BALB C , Mice, Nude , Mucin-1/biosynthesis , Mucin-1/metabolism , Peptides/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/biosynthesis , Quinazolines/pharmacology , RNA Interference , RNA, Small Interfering , Receptor, ErbB-2/biosynthesis , Trastuzumab , Xenograft Model Antitumor Assays
8.
Ann Oncol ; 25(4): 773-780, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24351401

ABSTRACT

Insights into tumour biology of breast cancer have led the path towards the introduction of targeted treatment approaches; still, breast cancer-related mortality remains relatively high. Efforts in the field of basic research revealed new druggable targets which now await validation within the context of clinical trials. Therefore, questions concerning the optimal design of future studies are becoming even more pertinent. Aspects such as the ideal end point, availability of predictive markers to identify the optimal cohort for drug testing, or potential mechanisms of resistance need to be resolved. An expert panel representing the academic community, the pharmaceutical industry, as well as European Regulatory Authorities met in Vienna, Austria, in November 2012, in order to discuss breast cancer biology, identification of novel biological targets and optimal drug development with the aim of treatment individualization. This article summarizes statements and perspectives provided by the meeting participants.


Subject(s)
Receptor, ErbB-2/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy , Clinical Trials as Topic , Female , Humans , Molecular Targeted Therapy , Signal Transduction , Triple Negative Breast Neoplasms/classification , Triple Negative Breast Neoplasms/genetics
9.
Oncogene ; 30(22): 2547-57, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21278786

ABSTRACT

There is a strong rationale to therapeutically target the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway in breast cancer since it is highly deregulated in this disease and it also mediates resistance to anti-HER2 therapies. However, initial studies with rapalogs, allosteric inhibitors of mTORC1, have resulted in limited clinical efficacy probably due to the release of a negative regulatory feedback loop that triggers AKT and ERK signaling. Since activation of AKT occurs via PI3K, we decided to explore whether PI3K inhibitors prevent the activation of these compensatory pathways. Using HER2-overexpressing breast cancer cells as a model, we observed that PI3K inhibitors abolished AKT activation. However, PI3K inhibition resulted in a compensatory activation of the ERK signaling pathway. This enhanced ERK signaling occurred as a result of activation of HER family receptors as evidenced by induction of HER receptors dimerization and phosphorylation, increased expression of HER3 and binding of adaptor molecules to HER2 and HER3. The activation of ERK was prevented with either MEK inhibitors or anti-HER2 monoclonal antibodies and tyrosine kinase inhibitors. Combined administration of PI3K inhibitors with either HER2 or MEK inhibitors resulted in decreased proliferation, enhanced cell death and superior anti-tumor activity compared with single agent PI3K inhibitors. Our findings indicate that PI3K inhibition in HER2-overexpressing breast cancer activates a new compensatory pathway that results in ERK dependency. Combined anti-MEK or anti-HER2 therapy with PI3K inhibitors may be required in order to achieve optimal efficacy in HER2-overexpressing breast cancer. This approach warrants clinical evaluation.


Subject(s)
Breast Neoplasms/drug therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/metabolism , Breast Neoplasms/enzymology , Cell Line, Tumor , Female , Humans , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-3/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors
10.
Oncogene ; 29(3): 325-34, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19855434

ABSTRACT

The anti-HER2 antibody Trastuzumab (Herceptin) has been proven to be effective in the treatment of HER2-overexpressing breast cancer; resistance, however, invariably emerges in metastatic tumors. The expression of p95-HER2, a form of HER2 with a truncated extracellular domain that lacks the Trastuzumab binding epitope, has been implicated as a mechanism of resistance to the antibody. We utilized an in vivo tumor model that overexpresses p95-HER2 and showed it to be resistant to the signaling and antitumor effects of Trastuzumab. We find that both full-length and p95-HER2 interact with the HSP90 chaperone protein and are degraded in tumor cells exposed to HSP90 inhibitors in tissue culture and in vivo. Loss of expression of p95-HER2 is accompanied by downregulation of the phosphoinositide-3 kinase/AKT and extracellular signal-regulated kinase signaling pathways and inhibition of cell proliferation. Chronic administration of HSP90 inhibitors in vivo results in sustained loss of HER2 and p95-HER2 expression and inhibition of AKT activation, together with induction of apoptosis and complete inhibition of tumor growth in Trastuzumab-resistant, p95-HER2-overexpressing models. Thus, p95-HER2 is an HSP90 client protein, the expression and function of which can be effectively suppressed in vivo by HSP90 inhibitors. HSP90 inhibition is therefore a potentially effective therapeutic strategy for p95-HER2-mediated Trastuzumab-resistant breast cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Neoplasms/drug therapy , Receptor, ErbB-2/metabolism , 3T3 Cells , Animals , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Binding Sites/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Immunoblotting , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Trastuzumab , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Oncogene ; 28(6): 803-14, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19060928

ABSTRACT

Lapatinib is a human epidermal growth factor receptor 2 (HER2) tyrosine kinase inhibitor (TKI) that has clinical activity in HER2-amplified breast cancer. In vitro studies have shown that lapatinib enhances the effects of the monoclonal antibody trastuzumab suggesting partially non-overlapping mechanisms of action. To dissect these mechanisms, we have studied the effects of lapatinib and trastuzumab on receptor expression and receptor signaling and have identified a new potential mechanism for the enhanced antitumor activity of the combination. Lapatinib, given alone or in combination with trastuzumab to HER2-overexpressing breast cancer cells SKBR3 and MCF7-HER2, inhibited HER2 phosphorylation, prevented receptor ubiquitination and resulted in a marked accumulation of inactive receptors at the cell surface. By contrast, trastuzumab alone caused enhanced HER2 phosphorylation, ubiquitination and degradation of the receptor. By immunoprecipitation and computational protein modeling techniques we have shown that the lapatinib-induced HER2 accumulation at the cell surface also results in the stabilization of inactive HER2 homo- (HER2/HER2) and hetero- (HER2/EGFR and HER2/HER3) dimers. Lapatinib-induced accumulation of HER2 and trastuzumab-mediated downregulation of HER2 was also observed in vivo, where the combination of the two agents triggered complete tumor remissions in all cases after 10 days of treatment. Accumulation of HER2 at the cell surface by lapatinib enhanced immune-mediated trastuzumab-dependent cytotoxicity. We propose that this is a novel mechanism of action of the combination that may be clinically relevant and exploitable in the therapy of patients with HER2-positive tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Breast Neoplasms/drug therapy , Drug Synergism , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lapatinib , Mice , Neoplasm Transplantation , Remission Induction , Signal Transduction , Trastuzumab
12.
Br J Cancer ; 99(10): 1607-12, 2008 Nov 18.
Article in English | MEDLINE | ID: mdl-18941461

ABSTRACT

Around 15% of colorectal cancers (CRCs) show microsatellite instability (MSI) due to dysfunction of the mismatch repair system (MMR). As a consequence of this, MSI tumours tend to accumulate errors in mononucleotide repeats as those in genes implicated in repairing double-strand breaks (DSBs). Previous studies have shown that irinotecan (CPT-11), a chemotherapy agent inducing DSB, is more active in MSI than in microsatellite stable (MSS) CRC. The purpose of this study was to compare the sensitivity to CPT-11 in a series of CRC cell lines with either proficient or deficient MMR and to assess the mutational status of two DSB repair genes, MRE11 and RAD50, in these cell lines. hMLH1-deficient cell lines due to either epigenetic silencing or mutation showed very similar IC(50) and were four- to nine-fold more sensitive to CPT-11 than the MSS line. Cell lines harbouring mutations in both MRE11 and RAD50 were most sensitive to CPT-11. We conclude that MSI cell lines display higher sensitivity to CPT-11 than MSS cells. Mutation of MRE11 and RAD50 could account for this difference in response to CPT-11. Future clinical trials tailoring chemotherapy regimens based on microsatellite status are warranted.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , DNA Repair Enzymes/genetics , Microsatellite Instability , Nuclear Proteins/genetics , Acid Anhydride Hydrolases , Camptothecin/pharmacology , Cell Line, Tumor/drug effects , Cytotoxins/pharmacology , DNA-Binding Proteins/genetics , Humans , Irinotecan , MRE11 Homologue Protein , MutL Protein Homolog 1
14.
Br J Cancer ; 91(10): 1842-50, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15494717

ABSTRACT

Expression of the castration-induced clusterin protein is incompatible with the survival of human prostate cancer cells in tissues and in cell culture. To investigate the fate of human prostate epithelial cells, when engineered to maintain expression of clusterin protein, we have used an IRES-hyg vector and hygromycin selection. PC-3 prostate tumour cells were substantially more sensitive to clusterin expression than nonmalignant PNT1a cells, showing multiple phenotypic changes including cell cycle arrest and increased apoptosis. The results strengthen the hypothesis that clusterin expression is proapoptotic. Expression of exogenous clusterin in both cell types resulted in its relocation from the cytoplasm and a nuclear accumulation of the protein, as was also seen in the same cells when apoptosis was induced by etoposide treatment. To survive clusterin expression, the PC-3 tumour cells developed apoptosis-inhibitory properties. This could have significance for the resistance of prostate cancers to chemo/radiotherapy, where clusterin overexpression is observed.


Subject(s)
Apoptosis , Glycoproteins/metabolism , Molecular Chaperones/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Cell Cycle , Cell Line, Transformed , Cell Proliferation , Clone Cells , Clusterin , Epithelial Cells/metabolism , Humans , Male , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Prostate/cytology , Prostatic Neoplasms/pathology , Transfection , Tumor Cells, Cultured
15.
Eur J Endocrinol ; 150(4): 591-603, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15080791

ABSTRACT

OBJECTIVE: Calcitriol analogues might represent an interesting new therapy for benign prostate hyperplasia (BPH). We here report the preclinical characterization of BXL-628, an analogue selected for an ongoing double-blind, randomized, placebo-controlled phase II trial in BPH. DESIGN: Experiments with BXL-628 were carried out in human BPH cells and in the ventral prostate of intact and castrated rats. METHODS: BPH cell and rat prostate growth were evaluated along with morphological and biochemical hallmarks of apoptosis. RESULTS: BXL-628 inhibited human BPH cell proliferation and induced apoptosis even in the presence of androgens or growth factors. It also decreased prostate growth to an extent similar to finasteride, inducing DNA fragmentation and apoptosis, both in intact and in testosterone-supplemented castrated rats. Accordingly, BXL-628, like finasteride, increased the expression of clusterin, a prostatic atrophy marker. However, BXL-628 did not inhibit 5 alpha-reductase 1 and 2, did not bind to the androgen receptor (AR) in BPH homogenates and did not affect AR-coupled luciferase activity. In addition, BXL-628 did not affect rat pituitary and testis activity or calcemia. CONCLUSIONS: BXL-628 inhibited in vitro and in vivo prostate cell proliferation, and therefore might represent a novel, interesting option for the treatment of BPH.


Subject(s)
Calcitriol/analogs & derivatives , Calcitriol/administration & dosage , Prostate/drug effects , Prostatic Hyperplasia/drug therapy , Animals , Cell Division/drug effects , Clinical Trials, Phase II as Topic , Drug Evaluation, Preclinical , Humans , Male , Orchiectomy , Prostate/pathology , Prostatic Hyperplasia/pathology , Randomized Controlled Trials as Topic , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured/drug effects
16.
Endocrinology ; 144(7): 3046-57, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12810561

ABSTRACT

We have recently found that analog V (BXL-353, a calcitriol analog) inhibits growth factor (GF)-stimulated human benign prostate hyperplasia (BPH) cell proliferation by disrupting signal transduction, reducing Bcl-2 expression, and inducing apoptosis. We now report that BXL-353 blocks in vitro and in vivo testosterone (T) activity. BPH cells responded to T and dihydrotestosterone (DHT) with dose-dependent growth and reduced apoptosis. Exposure of BPH cells to BXL-353 significantly antagonized both T- and DHT-induced proliferation and induced apoptosis, even in the presence of T. To verify whether BXL-353 reduced prostate growth in vivo, we administered it orally to either intact or castrated rats, supplemented with T enanthate. Nonhypercalcemic doses of BXL-353 time- and dose-dependently reduced the androgen effect on ventral prostate weight, similarly to finasteride. Comparable results were obtained after chronic administration of BXL-353 to intact rats. Clusterin (an atrophy marker) gene and protein were up-regulated by BXL-353 in rat prostate, and nuclear fragmentation was widely present. The antiandrogenic properties of BXL-353 did not interfere with pituitary and testis function, as assessed by serum determination of rat LH and T. BXL-353 did not compete for androgen binding to BPH homogenates and failed to inhibit 5alpha-reductase type 1 and type 2 activities. In conclusion, BXL-353 blocks in vitro and in vivo androgen-stimulated prostate cell growth, probably acting downstream from the androgen receptor, without affecting calcemia or sex hormone secretion. BXL-353 and other vitamin D(3) analogs might thus represent an interesting class of compounds for treating patients with BPH.


Subject(s)
Calcitriol/analogs & derivatives , Gonadal Steroid Hormones/pharmacology , Prostate/pathology , Prostatic Hyperplasia/drug therapy , Testosterone/pharmacology , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Aging/pathology , Androgen Antagonists/pharmacology , Animals , Apoptosis/drug effects , Atrophy , CHO Cells , Clusterin , Cricetinae , Dihydrotestosterone/pharmacology , Gene Expression/drug effects , Glycoproteins/genetics , Glycoproteins/metabolism , Gonadal Steroid Hormones/blood , Humans , Luteinizing Hormone/blood , Male , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Prostate/drug effects , Prostatic Hyperplasia/pathology , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Androgen/genetics , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Stromal Cells/cytology , Stromal Cells/drug effects , Testosterone/blood , Up-Regulation/drug effects
17.
Ann N Y Acad Sci ; 1010: 514-9, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15033782

ABSTRACT

Clusterin gene expression is potently induced in experimental models in which apoptosis is activated, such as rat prostate involution following castration. Nevertheless, its precise physiological role has not yet been established, and both anti-apoptotic and pro-apoptotic functions have been suggested for this gene. Clusterin expression level depends on cell proliferation state, and we recently showed that its over-expression inhibited cell cycle progression of SV40-immortalized human prostate epithelial cells PNT2 and PNT1a. Here we studied clusterin expression in PNT1a cells subjected to serum-starvation with the aim of defining clusterin early molecular changes following apoptosis induction. Under serum-starvation conditions, decreased growth rate, slow rounding-up of cells, cell detachment, and formation of apoptotic bodies indicative of anoikis (detachment-induced apoptosis) were preceded by significant downregulation of 70 kDa clusterin precursor and upregulation of 45-40 kDa isoforms. On the 8th day of serum-free culturing, only the higher molecular weight protein-band of about 45 kDa was clearly induced and accumulated in detached cells and apoptotic bodies in which PARP was activated. Anoikis was preceded by induction and transloction of a 45-kDa clusterin isoform to the nucleus. Thus, nuclear targeting of a specific 45-kDa isoform of clusterin appeared to be an early and specific molecular signal triggering anoikis-death. Considering also that clusterin is downregulated during prostate cancer onset and progression, and that its upregulation has inhibited DNA synthesis and cell cycle progression of immortalized human prostate epithelial cells, we suggest that clusterin might be a new anti-oncogene in the prostate.


Subject(s)
Active Transport, Cell Nucleus/physiology , Apoptosis/physiology , Glycoproteins/genetics , Molecular Chaperones/genetics , Simian virus 40/genetics , Cell Division , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Transformation, Viral , Clusterin , Culture Media, Serum-Free , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , Humans , Keratins/metabolism , Kinetics , Male , Molecular Chaperones/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...