Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 26(18): 4933-4946, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32576626

ABSTRACT

PURPOSE: Advances in prostate cancer lag behind other tumor types partly due to the paucity of models reflecting key milestones in prostate cancer progression. Therefore, we develop clinically relevant prostate cancer models. EXPERIMENTAL DESIGN: Since 1996, we have generated clinically annotated patient-derived xenografts (PDXs; the MDA PCa PDX series) linked to specific phenotypes reflecting all aspects of clinical prostate cancer. RESULTS: We studied two cell line-derived xenografts and the first 80 PDXs derived from 47 human prostate cancer donors. Of these, 47 PDXs derived from 22 donors are working models and can be expanded either as cell lines (MDA PCa 2a and 2b) or PDXs. The histopathologic, genomic, and molecular characteristics (androgen receptor, ERG, and PTEN loss) maintain fidelity with the human tumor and correlate with published findings. PDX growth response to mouse castration and targeted therapy illustrate their clinical utility. Comparative genomic hybridization and sequencing show significant differences in oncogenic pathways in pairs of PDXs derived from different areas of the same tumor. We also identified a recurrent focal deletion in an area that includes the speckle-type POZ protein-like (SPOPL) gene in PDXs derived from seven human donors of 28 studied (25%). SPOPL is a SPOP paralog, and SPOP mutations define a molecular subclass of prostate cancer. SPOPL deletions are found in 7% of The Cancer Genome Atlas prostate cancers, which suggests that our cohort is a reliable platform for targeted drug development. CONCLUSIONS: The MDA PCa PDX series is a dynamic resource that captures the molecular landscape of prostate cancers progressing under novel treatments and enables optimization of prostate cancer-specific, marker-driven therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Precision Medicine/methods , Prostatic Neoplasms/drug therapy , Adaptor Proteins, Vesicular Transport/genetics , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Comparative Genomic Hybridization , DNA Copy Number Variations , Humans , Male , Mice , Primary Cell Culture , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Sequence Deletion , Xenograft Model Antitumor Assays/methods
2.
Sci Rep ; 6: 35354, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27748439

ABSTRACT

Cytochrome P450 17α-hydroxylase/17,20-lyase (CYP17A1) is a validated treatment target for the treatment of metastatic castration-resistant prostate cancer (CRPC). Abiraterone acetate (AA) inhibits both 17α-hydroxylase (hydroxylase) and 17,20-lyase (lyase) reactions catalyzed by CYP17A1 and thus depletes androgen biosynthesis. However, coadministration of prednisone is required to suppress the mineralocorticoid excess and cortisol depletion that result from hydroxylase inhibition. VT-464, a nonsteroidal small molecule, selectively inhibits CYP17A1 lyase and therefore does not require prednisone supplementation. Administration of VT-464 in a metastatic CRPC patient presenting with high tumoral expression of both androgen receptor (AR) and CYP17A1, showed significant reduction in the level of both dehydroepiandrosterone (DHEA) and serum PSA. Treatment of a CRPC patient-derived xenograft, MDA-PCa-133 expressing H874Y AR mutant with VT-464, reduced the increase in tumor volume in castrate male mice more than twice as much as the vehicle (P < 0.05). Mass spectrometry analysis of post-treatment xenograft tumor tissues showed that VT-464 significantly decreased intratumoral androgens but not cortisol. VT-464 also reduced AR signaling more effectively than abiraterone in cultured PCa cells expressing T877A AR mutant. Collectively, this study suggests that VT-464 therapy can effectively treat CRPC and be used in precision medicine based on androgen receptor mutation status.


Subject(s)
Naphthalenes/administration & dosage , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Triazoles/administration & dosage , Abiraterone Acetate/administration & dosage , Androgens/biosynthesis , Animals , Biopsy , Cell Line, Tumor , Dehydroepiandrosterone/chemistry , Humans , Hydrocortisone/blood , Male , Mass Spectrometry , Mice , Mice, SCID , Neoplasm Transplantation , Precision Medicine , Prednisone/administration & dosage , Receptors, Androgen/genetics , Signal Transduction , Steroid 17-alpha-Hydroxylase/metabolism
3.
EBioMedicine ; 7: 85-93, 2016 May.
Article in English | MEDLINE | ID: mdl-27322462

ABSTRACT

BACKGROUND: In the Prostate Cancer Prevention Trial, finasteride selectively suppressed low-grade prostate cancer and significantly reduced the incidence of prostate cancer in men treated with finasteride compared with placebo. However, an apparent increase in high-grade disease was also observed among men randomized to finasteride. We aimed to determine why and hypothesized that there is a grade-dependent response to finasteride. METHODS: From 2007 to 2012, we randomized dynamically by intranet-accessible software 183 men with localized prostate cancer to receive 5mg finasteride or placebo daily in a double-blind study during the 4-6weeks preceding prostatectomy. As the primary end point, the expression of a predefined molecular signature (ERß, UBE2C, SRD5A2, and VEGF) differentiating high- and low-grade tumors in Gleason grade (GG) 3 areas of finasteride-exposed tumors from those in GG3 areas of placebo-exposed tumors, adjusted for Gleason score (GS) at prostatectomy, was compared. We also determined androgen receptor (AR) levels, Ki-67, and cleaved caspase 3 to evaluate the effects of finasteride on the expression of its downstream target, cell proliferation, and apoptosis, respectively. The expression of these markers was also compared across grades between and within treatment groups. Logistic regression was used to assess the expression of markers. FINDINGS: We found that the predetermined molecular signature did not distinguish GG3 from GG4 areas in the placebo group. However, AR expression was significantly lower in the GG4 areas of the finasteride group than in those of the placebo group. Within the finasteride group, AR expression was also lower in GG4 than in GG3 areas, but not significantly. Expression of cleaved caspase 3 was significantly increased in both GG3 and GG4 areas in the finasteride group compared to the placebo group, although it was lower in GG4 than in GG3 areas in both groups. INTERPRETATION: We showed that finasteride's effect on apoptosis and AR expression is tumor grade dependent after short-term intervention. This may explain finasteride's selective suppression of low-grade tumors observed in the PCPT.


Subject(s)
Finasteride/administration & dosage , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Administration, Oral , Aged , Apoptosis , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Double-Blind Method , Finasteride/pharmacology , Humans , Logistic Models , Male , Middle Aged , Neoplasm Grading , Prostatic Neoplasms/metabolism , Treatment Outcome
4.
Nat Commun ; 7: 11674, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27241286

ABSTRACT

White adipose tissue (WAT) overgrowth in obesity is linked with increased aggressiveness of certain cancers. Adipose stromal cells (ASCs) can become mobilized from WAT, recruited by tumours and promote cancer progression. Mechanisms underlying ASC trafficking are unclear. Here we demonstrate that chemokines CXCL1 and CXCL8 chemoattract ASC by signalling through their receptors, CXCR1 and CXCR2, in cell culture models. We further show that obese patients with prostate cancer have increased epithelial CXCL1 expression. Concomitantly, we observe that cells with ASC phenotype are mobilized and infiltrate tumours in obese patients. Using mouse models, we show that the CXCL1 chemokine gradient is required for the obesity-dependent tumour ASC recruitment, vascularization and tumour growth promotion. We demonstrate that αSMA expression in ASCs is induced by chemokine signalling and mediates the stimulatory effects of ASCs on endothelial cells. Our data suggest that ASC recruitment to tumours, driven by CXCL1 and CXCL8, promotes prostate cancer progression.


Subject(s)
Cell Movement/physiology , Chemokine CXCL1/metabolism , Mesenchymal Stem Cells/pathology , Obesity/pathology , Prostatic Neoplasms/pathology , Tumor Microenvironment/physiology , Actins/metabolism , Adipocytes/pathology , Adipose Tissue, White/cytology , Adipose Tissue, White/pathology , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Chemokine CXCL1/genetics , Diet, High-Fat/adverse effects , Disease Progression , Endothelial Cells/pathology , Humans , Interleukin-8/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Middle Aged , Neovascularization, Pathologic/pathology , Obesity/complications , Obesity/metabolism , Primary Cell Culture , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/complications , RNA, Small Interfering/metabolism , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Tissue Array Analysis , Xenograft Model Antitumor Assays
5.
Cancer Discov ; 6(1): 80-95, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26701088

ABSTRACT

UNLABELLED: The signaling mechanisms between prostate cancer cells and infiltrating immune cells may illuminate novel therapeutic approaches. Here, utilizing a prostate adenocarcinoma model driven by loss of Pten and Smad4, we identify polymorphonuclear myeloid-derived suppressor cells (MDSC) as the major infiltrating immune cell type, and depletion of MDSCs blocks progression. Employing a novel dual reporter prostate cancer model, epithelial and stromal transcriptomic profiling identified CXCL5 as a cancer-secreted chemokine to attract CXCR2-expressing MDSCs, and, correspondingly, pharmacologic inhibition of CXCR2 impeded tumor progression. Integrated analyses identified hyperactivated Hippo-YAP signaling in driving CXCL5 upregulation in cancer cells through the YAP-TEAD complex and promoting MDSC recruitment. Clinicopathologic studies reveal upregulation and activation of YAP1 in a subset of human prostate tumors, and the YAP1 signature is enriched in primary prostate tumor samples with stronger expression of MDSC-relevant genes. Together, YAP-driven MDSC recruitment via heterotypic CXCL5-CXCR2 signaling reveals an effective therapeutic strategy for advanced prostate cancer. SIGNIFICANCE: We demonstrate a critical role of MDSCs in prostate tumor progression and discover a cancer cell nonautonomous function of the Hippo-YAP pathway in regulation of CXCL5, a ligand for CXCR2-expressing MDSCs. Pharmacologic elimination of MDSCs or blocking the heterotypic CXCL5-CXCR2 signaling circuit elicits robust antitumor responses and prolongs survival.


Subject(s)
Chemokine CXCL5/genetics , Myeloid Cells/immunology , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/immunology , Smad4 Protein/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Chemokine CXCL5/metabolism , Disease Progression , Hippo Signaling Pathway , Humans , Male , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Signal Transduction , Transcription Factors , YAP-Signaling Proteins
6.
Sci Transl Med ; 6(252): 252ra122, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25186177

ABSTRACT

Bone is the most common site of prostate cancer (PCa) progression to a therapy-resistant, lethal phenotype. We found that blockade of fibroblast growth factor receptors (FGFRs) with the receptor tyrosine kinase inhibitor dovitinib has clinical activity in a subset of men with castration-resistant PCa and bone metastases. Our integrated analyses suggest that FGF signaling mediates a positive feedback loop between PCa cells and bone cells and that blockade of FGFR1 in osteoblasts partially mediates the antitumor activity of dovitinib by improving bone quality and by blocking PCa cell-bone cell interaction. These findings account for clinical observations such as reductions in lesion size and intensity on bone scans, lymph node size, and tumor-specific symptoms without proportional declines in serum prostate-specific antigen concentration. Our findings suggest that targeting FGFR has therapeutic activity in advanced PCa and provide direction for the development of therapies with FGFR inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Prostatic Neoplasms/pathology , Quinolones/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Benzimidazoles/pharmacology , Bone Neoplasms/pathology , Bone and Bones/drug effects , Bone and Bones/metabolism , Cell Line, Tumor , Disease Models, Animal , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Osteoblasts/drug effects , Osteoblasts/metabolism , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Quinolones/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Stromal Cells/drug effects , Stromal Cells/pathology , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
7.
Sci Signal ; 7(326): ra47, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24847116

ABSTRACT

Androgen deprivation is the standard treatment for advanced prostate cancer (PCa), but most patients ultimately develop resistance and tumor recurrence. We found that MYB is transcriptionally activated by androgen deprivation therapy or genetic silencing of the androgen receptor (AR). MYB silencing inhibited PCa growth in culture and xenografts in mice. Microarray data revealed that c-Myb and AR shared a subset of target genes that encode DNA damage response (DDR) proteins, suggesting that c-Myb may supplant AR as the dominant regulator of their common DDR target genes in AR inhibition-resistant or AR-negative PCa. Gene signatures including AR, MYB, and their common DDR-associated target genes positively correlated with metastasis, castration resistance, tumor recurrence, and decreased survival in PCa patients. In culture and in xenograft-bearing mice, a combination strategy involving the knockdown of MYB, BRCA1, or TOPBP1 or the abrogation of cell cycle checkpoint arrest with AZD7762, an inhibitor of the checkpoint kinase Chk1, increased the cytotoxicity of the poly[adenosine 5'-diphosphate (ADP)-ribose] polymerase (PARP) inhibitor olaparib in PCa cells. Our results reveal new mechanism-based therapeutic approaches for PCa by targeting PARP and the DDR pathway involving c-Myb, TopBP1, ataxia telangiectasia mutated- and Rad3-related (ATR), and Chk1.


Subject(s)
DNA Damage , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Thiophenes/pharmacology , Urea/analogs & derivatives , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Castration , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Male , Mice , Mice, Nude , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Urea/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...