Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nature ; 471(7339): 523-6, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21430781

ABSTRACT

Human lung adenocarcinomas with activating mutations in EGFR (epidermal growth factor receptor) often respond to treatment with EGFR tyrosine kinase inhibitors (TKIs), but the magnitude of tumour regression is variable and transient. This heterogeneity in treatment response could result from genetic modifiers that regulate the degree to which tumour cells are dependent on mutant EGFR. Through a pooled RNA interference screen, we show that knockdown of FAS and several components of the NF-κB pathway specifically enhanced cell death induced by the EGFR TKI erlotinib in EGFR-mutant lung cancer cells. Activation of NF-κB through overexpression of c-FLIP or IKK (also known as CFLAR and IKBKB, respectively), or silencing of IκB (also known as NFKBIA), rescued EGFR-mutant lung cancer cells from EGFR TKI treatment. Genetic or pharmacologic inhibition of NF-κB enhanced erlotinib-induced apoptosis in erlotinib-sensitive and erlotinib-resistant EGFR-mutant lung cancer models. Increased expression of the NF-κB inhibitor IκB predicted for improved response and survival in EGFR-mutant lung cancer patients treated with EGFR TKI. These data identify NF-κB as a potential companion drug target, together with EGFR, in EGFR-mutant lung cancers and provide insight into the mechanisms by which tumour cells escape from oncogene dependence.


Subject(s)
ErbB Receptors/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutant Proteins/metabolism , NF-kappa B/metabolism , Signal Transduction , fas Receptor/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Down-Regulation , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Erlotinib Hydrochloride , Genes, erbB-1/genetics , Humans , I-kappa B Proteins/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Models, Biological , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Mutation/genetics , NF-kappa B/antagonists & inhibitors , Quinazolines/pharmacology , Quinazolines/therapeutic use , RNA Interference , Signal Transduction/drug effects , fas Receptor/antagonists & inhibitors
2.
Proc Natl Acad Sci U S A ; 111(30): 11139-44, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25024180

ABSTRACT

Primary prostate cancer is the most common malignancy in men but has highly variable outcomes, highlighting the need for biomarkers to determine which patients can be managed conservatively. Few large prostate oncogenome resources currently exist that combine the molecular and clinical outcome data necessary to discover prognostic biomarkers. Previously, we found an association between relapse and the pattern of DNA copy number alteration (CNA) in 168 primary tumors, raising the possibility of CNA as a prognostic biomarker. Here we examine this question by profiling an additional 104 primary prostate cancers and updating the initial 168 patient cohort with long-term clinical outcome. We find that CNA burden across the genome, defined as the percentage of the tumor genome affected by CNA, was associated with biochemical recurrence and metastasis after surgery in these two cohorts, independent of the prostate-specific antigen biomarker or Gleason grade, a major existing histopathological prognostic variable in prostate cancer. Moreover, CNA burden was associated with biochemical recurrence in intermediate-risk Gleason 7 prostate cancers, independent of prostate-specific antigen or nomogram score. We further demonstrate that CNA burden can be measured in diagnostic needle biopsies using low-input whole-genome sequencing, setting the stage for studies of prognostic impact in conservatively treated cohorts.


Subject(s)
Biomarkers, Tumor/genetics , DNA Copy Number Variations , DNA, Neoplasm/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Biopsy, Needle , Disease-Free Survival , Follow-Up Studies , Humans , Male , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Survival Rate
3.
Nature ; 468(7326): 968-72, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21107320

ABSTRACT

Oncogenic mutations in the serine/threonine kinase B-RAF (also known as BRAF) are found in 50-70% of malignant melanomas. Pre-clinical studies have demonstrated that the B-RAF(V600E) mutation predicts a dependency on the mitogen-activated protein kinase (MAPK) signalling cascade in melanoma-an observation that has been validated by the success of RAF and MEK inhibitors in clinical trials. However, clinical responses to targeted anticancer therapeutics are frequently confounded by de novo or acquired resistance. Identification of resistance mechanisms in a manner that elucidates alternative 'druggable' targets may inform effective long-term treatment strategies. Here we expressed ∼600 kinase and kinase-related open reading frames (ORFs) in parallel to interrogate resistance to a selective RAF kinase inhibitor. We identified MAP3K8 (the gene encoding COT/Tpl2) as a MAPK pathway agonist that drives resistance to RAF inhibition in B-RAF(V600E) cell lines. COT activates ERK primarily through MEK-dependent mechanisms that do not require RAF signalling. Moreover, COT expression is associated with de novo resistance in B-RAF(V600E) cultured cell lines and acquired resistance in melanoma cells and tissue obtained from relapsing patients following treatment with MEK or RAF inhibitors. We further identify combinatorial MAPK pathway inhibition or targeting of COT kinase activity as possible therapeutic strategies for reducing MAPK pathway activation in this setting. Together, these results provide new insights into resistance mechanisms involving the MAPK pathway and articulate an integrative approach through which high-throughput functional screens may inform the development of novel therapeutic strategies.


Subject(s)
Drug Resistance, Neoplasm , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Allosteric Regulation , Cell Line, Tumor , Clinical Trials as Topic , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Activation/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Library , Humans , Indoles/pharmacology , Indoles/therapeutic use , MAP Kinase Kinase Kinases/genetics , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/genetics , Melanoma/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Open Reading Frames/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Vemurafenib
4.
Nat Methods ; 8(8): 659-61, 2011 Jun 26.
Article in English | MEDLINE | ID: mdl-21706014

ABSTRACT

Functional characterization of the human genome requires tools for systematically modulating gene expression in both loss-of-function and gain-of-function experiments. We describe the production of a sequence-confirmed, clonal collection of over 16,100 human open-reading frames (ORFs) encoded in a versatile Gateway vector system. Using this ORFeome resource, we created a genome-scale expression collection in a lentiviral vector, thereby enabling both targeted experiments and high-throughput screens in diverse cell types.


Subject(s)
Cloning, Molecular/methods , Genetic Vectors/genetics , Genomic Library , Lentivirus/genetics , Humans , Open Reading Frames
5.
Cancer Cell ; 10(4): 321-30, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17010675

ABSTRACT

Although androgen receptor (AR)-mediated signaling is central to prostate cancer, the ability to modulate AR signaling states is limited. Here we establish a chemical genomic approach for discovery and target prediction of modulators of cancer phenotypes, as exemplified by AR signaling. We first identify AR activation inhibitors, including a group of structurally related compounds comprising celastrol, gedunin, and derivatives. To develop an in silico approach for target pathway identification, we apply a gene expression-based analysis that classifies HSP90 inhibitors as having similar activity to celastrol and gedunin. Validating this prediction, we demonstrate that celastrol and gedunin inhibit HSP90 activity and HSP90 clients, including AR. Broadly, this work identifies new modes of HSP90 modulation through a gene expression-based strategy.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression/drug effects , Genome, Human , HSP90 Heat-Shock Proteins/metabolism , Receptors, Androgen/metabolism , Antibiotics, Antineoplastic/pharmacology , Benzoquinones/pharmacology , Cell Culture Techniques , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , ErbB Receptors/metabolism , Fusion Proteins, bcr-abl/metabolism , Gene Expression Profiling , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Inhibitory Concentration 50 , Lactams, Macrocyclic/pharmacology , Limonins/pharmacology , Male , Metribolone/pharmacology , Pentacyclic Triterpenes , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/analysis , Reproducibility of Results , Triterpenes/pharmacology , fms-Like Tyrosine Kinase 3/metabolism
6.
Nat Genet ; 33(2): 155-61, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12524544

ABSTRACT

Nuclear export of mRNA is mediated by a complex machinery of RNA-binding proteins that recognizes and routes mRNAs through a messenger ribonucleoprotein (mRNP) network. The full spectrum of mRNA cargoes for any dedicated mRNA export factor is unknown. We identified the mRNAs that bind two conserved yeast mRNA export factors, Yra1 (refs. 1-5) and Mex67 (refs. 6,7), on a genome-wide scale and determined their level of binding. Yra1 and Mex67 bind approximately 1,000 and 1,150 mRNAs, respectively, corresponding to almost 20% of the yeast genome and roughly 36% of all transcriptional events each. The binding level of Yra1 targets is related to their transcriptional frequency, but that of Mex67 targets is not. Yra1-bound transcripts are enriched in mRNAs that are regulated by a number of transcription factors. Yra1- and Mex67-bound populations also show enrichment of mRNAs encoding distinct functional classes of proteins, some of which are regulated by these transcription factors. We determined that one such transcription factor, Abf1 (refs. 8-10), associates with Yra1. These results indicate a previously unidentified specificity of mRNA export factors, which coordinates the export of transcriptionally co-regulated, functional classes of transcripts, perhaps through interactions with the transcriptional machinery.


Subject(s)
Active Transport, Cell Nucleus/physiology , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Profiling , Genome , In Situ Hybridization , Mutation , Nuclear Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Precipitin Tests , Protein Binding , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic
7.
Cancer Res ; 80(7): 1428-1437, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32015092

ABSTRACT

Genomic rearrangements leading to the aberrant expression of ERG are the most common early events in prostate cancer and are significantly enriched for the concomitant loss of PTEN. Genetically engineered mouse models reveal that ERG overexpression alone is not sufficient to induce tumorigenesis, but combined loss of PTEN results in an aggressive invasive phenotype. Here, we show that oncogenic ERG repressed PI3K signaling through direct transcriptional suppression of IRS2, leading to reduced RTK levels and activity. In accordance with this finding, ERG-positive human prostate cancers had a repressed AKT gene signature and transcriptional downregulation of IRS2. Although overexpression of IRS2 activated PI3K signaling, promoting cell migration in a PI3K-dependent manner, this did not fully recapitulate the phenotype seen with loss of PTEN as PI3K signaling is not as robust as observed in the setting of loss of PTEN. Importantly, deletions of the PTEN locus, which promotes active PI3K signaling, were among the most significant copy-number alterations that co-occurred with ERG genomic rearrangements. This work provides insight on how initiating oncogenic events may directly influence the selection of secondary concomitant alterations to promote oncogenic signaling during tumor evolution. SIGNIFICANCE: This work provides insight on how initiating oncogenic events may directly influence the selection of secondary concomitant alterations to promote tumorigenesis.


Subject(s)
Insulin Receptor Substrate Proteins/genetics , Oncogene Proteins/metabolism , Prostatic Neoplasms/genetics , Transcriptional Regulator ERG/metabolism , Animals , Carcinogenesis/genetics , Cell Line, Tumor , DNA Copy Number Variations , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Knockout Techniques , Gene Rearrangement , Humans , Insulin Receptor Substrate Proteins/metabolism , Male , Mice , Oncogene Proteins/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Promoter Regions, Genetic/genetics , Prostate/pathology , Prostatic Neoplasms/pathology , RNA-Seq , Signal Transduction/genetics , Transcriptional Regulator ERG/genetics , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 18(9): 1577-1586, 2019 09.
Article in English | MEDLINE | ID: mdl-31296553

ABSTRACT

On the basis of our previous work defining the molecular rationale for combined targeting of the PI3K and AR pathways in PTEN loss prostate cancer, the first clinical trial was recently reported demonstrating a significant benefit for combination therapy in patients with metastatic prostate cancer. In this phase II trial, loss of PTEN was a biomarker predictive of response to combined AKT and AR inhibition. Given that PTEN loss prostate cancers are significantly enriched for ERG genomic rearrangements, we evaluated how the aberrant expression of ERG may impact response to PI3K/AR-targeted therapy. Here, we show that overexpression of ERG in the setting of Pten loss promotes resistance to combined PI3K and AR pathway inhibition with associated maintenance of AR target gene expression. Importantly, following AR knockout in the setting of ERG overexpression, there is maintenance of a subset of AR lineage-specific target genes, making AR dispensable in this context. This has important clinical implications as even in the setting of the androgen-regulated TMPRSS2:ERG genomic rearrangement, ERG expression is never abolished following AR inhibition and may allow for cell survival following AR (lineage)-targeted therapies.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Animals , Benzamides , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Male , Mice, Knockout , Mice, SCID , Mice, Transgenic , Nitriles , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Quinolines/pharmacology , Receptors, Androgen/genetics , Signal Transduction/genetics , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism , Xenograft Model Antitumor Assays/methods
9.
Elife ; 72018 09 04.
Article in English | MEDLINE | ID: mdl-30178746

ABSTRACT

The level of copy number alteration (CNA), termed CNA burden, in the tumor genome is associated with recurrence of primary prostate cancer. Whether CNA burden is associated with prostate cancer survival or outcomes in other cancers is unknown. We analyzed the CNA landscape of conservatively treated prostate cancer in a biopsy and transurethral resection cohort, reflecting an increasingly common treatment approach. We find that CNA burden is prognostic for cancer-specific death, independent of standard clinical prognosticators. More broadly, we find CNA burden is significantly associated with disease-free and overall survival in primary breast, endometrial, renal clear cell, thyroid, and colorectal cancer in TCGA cohorts. To assess clinical applicability, we validated these findings in an independent pan-cancer cohort of patients whose tumors were sequenced using a clinically-certified next generation sequencing assay (MSK-IMPACT), where prognostic value varied based on cancer type. This prognostic association was affected by incorporating tumor purity in some cohorts. Overall, CNA burden of primary and metastatic tumors is a prognostic factor, potentially modulated by sample purity and measurable by current clinical sequencing.


Subject(s)
DNA Copy Number Variations/genetics , Neoplasm Recurrence, Local/genetics , Neoplasms/genetics , Neoplasms/pathology , Tumor Burden/genetics , Cell Death , Cohort Studies , Genome, Human , Humans , Kaplan-Meier Estimate , Prognosis , Exome Sequencing
10.
J Exp Med ; 215(1): 159-175, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29141866

ABSTRACT

Prostate-specific membrane antigen (PSMA) or folate hydrolase 1 (FOLH1) is highly expressed on prostate cancer. Its expression correlates inversely with survival and increases with tumor grade. However, the biological role of PSMA has not been explored, and its role in prostate cancer remained elusive. Filling this gap, we demonstrate that in prostate cancer, PSMA initiates signaling upstream of PI3K through G protein-coupled receptors, specifically via the metabotropic glutamate receptor (mGluR). PSMA's carboxypeptidase activity releases glutamate from vitamin B9 and other glutamated substrates, which activate mGluR I. Activated mGluR I subsequently induces activation of phosphoinositide 3-kinase (PI3K) through phosphorylation of p110ß independent of PTEN loss. The p110ß isoform of PI3K plays a particularly important role in the pathogenesis of prostate cancer, but the origin of its activation was so far unknown. PSMA expression correlated with PI3K-Akt signaling in cells, animal models, and patients. We interrogated the activity of the PSMA-PI3K axis through positron emission tomography and magnetic resonance imaging. Inhibition of PSMA in preclinical models inhibited PI3K signaling and promoted tumor regression. Our data present a novel oncogenic signaling role of PSMA that can be exploited for therapy and interrogated with imaging.

11.
Nat Commun ; 8(1): 1081, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29057879

ABSTRACT

A multigenic locus at 3p13-14, spanning FOXP1 to SHQ1, is commonly deleted in prostate cancer and lost broadly in a range of cancers but has unknown significance to oncogenesis or prognosis. Here, we report that FOXP1-SHQ1 deletion cooperates with PTEN loss to accelerate prostate oncogenesis and that loss of component genes correlates with prostate, breast, and head and neck cancer recurrence. We demonstrate that Foxp1-Shq1 deletion accelerates prostate tumorigenesis in mice in combination with Pten loss, consistent with the association of FOXP1-SHQ1 and PTEN loss observed in human cancers. Tumors with combined Foxp1-Shq1 and Pten deletion show increased proliferation and anaplastic dedifferentiation, as well as mTORC1 hyperactivation with reduced Akt phosphorylation. Foxp1-Shq1 deletion restores expression of AR target genes repressed in tumors with Pten loss, circumventing PI3K-mediated repression of the androgen axis. Moreover, FOXP1-SHQ1 deletion has prognostic relevance, with cancer recurrence associated with combined loss of PTEN and FOXP1-SHQ1 genes.


Subject(s)
Carrier Proteins/metabolism , Forkhead Transcription Factors/metabolism , PTEN Phosphohydrolase/metabolism , Repressor Proteins/metabolism , Animals , Carrier Proteins/genetics , Forkhead Transcription Factors/genetics , Humans , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/genetics
12.
Cell Rep ; 17(10): 2596-2606, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27926864

ABSTRACT

Inflammation is a risk factor for prostate cancer, but the mechanisms by which inflammation increases that risk are poorly understood. Here, we demonstrate that low expression of CD38 identifies a progenitor-like subset of luminal cells in the human prostate. CD38lo luminal cells are enriched in glands adjacent to inflammatory cells and exhibit epithelial nuclear factor κB (NF-κB) signaling. In response to oncogenic transformation, CD38lo luminal cells can initiate human prostate cancer in an in vivo tissue-regeneration assay. Finally, the CD38lo luminal phenotype and gene signature are associated with disease progression and poor outcome in prostate cancer. Our results suggest that prostate inflammation expands the pool of progenitor-like target cells susceptible to tumorigenesis.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Cell Transformation, Neoplastic/genetics , Inflammation/genetics , Prostatic Neoplasms/genetics , ADP-ribosyl Cyclase 1/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Male , NF-kappa B/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
13.
Mol Cancer Ther ; 14(1): 278-88, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25381262

ABSTRACT

Copy-number alterations (CNA) are among the most common molecular events in human prostate cancer genomes and are associated with worse prognosis. Identification of the oncogenic drivers within these CNAs is challenging due to the broad nature of these genomic gains or losses which can include large numbers of genes within a given region. Here, we profiled the genomes of four genetically engineered mouse prostate cancer models that reflect oncogenic events common in human prostate tumors, with the goal of integrating these data with human prostate cancer datasets to identify shared molecular events. Met was amplified in 67% of prostate tumors from Pten p53 prostate conditional null mice and in approximately 30% of metastatic human prostate cancer specimens, often in association with loss of PTEN and TP53. In murine tumors with Met amplification, Met copy-number gain and expression was present in some cells but not others, revealing intratumoral heterogeneity. Forced MET overexpression in non-MET-amplified prostate tumor cells activated PI3K and MAPK signaling and promoted cell proliferation and tumor growth, whereas MET kinase inhibition selectively impaired the growth of tumors with Met amplification. However, the impact of MET inhibitor therapy was compromised by the persistent growth of non-Met-amplified cells within Met-amplified tumors. These findings establish the importance of MET in prostate cancer progression but reveal potential limitations in the clinical use of MET inhibitors in late-stage prostate cancer.


Subject(s)
PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-met/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Copy Number Variations , Gene Amplification , Gene Expression Profiling , Genetic Heterogeneity , Genome , Humans , MAP Kinase Signaling System , Male , Mice , Mice, Transgenic , Neoplasms, Experimental , Protein Kinase Inhibitors/pharmacology
14.
Neoplasia ; 16(1): 14-20, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24563616

ABSTRACT

BACKGROUND: Recurrent mutations in the Speckle-Type POZ Protein (SPOP) gene occur in up to 15% of prostate cancers. However, the frequency and features of cancers with these mutations across different populations is unknown. OBJECTIVE: To investigate SPOP mutations across diverse cohorts and validate a series of assays employing high-resolution melting (HRM) analysis and Sanger sequencing for mutational analysis of formalin-fixed paraffin-embedded material. DESIGN SETTING AND PARTICIPANTS: 720 prostate cancer samples from six international cohorts spanning Caucasian, African American, and Asian patients, including both prostate-specific antigen-screened and unscreened populations, were screened for their SPOP mutation status. Status of SPOP was correlated to molecular features (ERG rearrangement, PTEN deletion, and CHD1 deletion) as well as clinical and pathologic features. RESULTS AND LIMITATIONS: Overall frequency of SPOP mutations was 8.1% (4.6% to 14.4%), SPOP mutation was inversely associated with ERG rearrangement (P<.01), and SPOP mutant (SPOPmut) cancers had higher rates of CHD1 deletions (P<.01). There were no significant differences in biochemical recurrence in SPOPmut cancers. Limitations of this study include missing mutational data due to sample quality and lack of power to identify a difference in clinical outcomes. CONCLUSION: SPOP is mutated in 4.6% to 14.4% of patients with prostate cancer across different ethnic and demographic backgrounds. There was no significant association between SPOP mutations with ethnicity, clinical, or pathologic parameters. Mutual exclusivity of SPOP mutation with ERG rearrangement as well as a high association with CHD1 deletion reinforces SPOP mutation as defining a distinct molecular subclass of prostate cancer.


Subject(s)
DNA Helicases/genetics , DNA-Binding Proteins/genetics , Mutation , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , Adult , Aged , Aged, 80 and over , Cohort Studies , DNA Mutational Analysis , Exons , Gene Deletion , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasms/genetics , Prostatectomy , Prostatic Neoplasms/ethnology , Sequence Analysis, DNA , Transcriptional Regulator ERG
16.
Cancer Discov ; 3(11): 1245-53, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24027196

ABSTRACT

UNLABELLED: We demonstrate that the androgen receptor (AR) regulates a transcriptional program of DNA repair genes that promotes prostate cancer radioresistance, providing a potential mechanism by which androgen deprivation therapy synergizes with ionizing radiation. Using a model of castration-resistant prostate cancer, we show that second-generation antiandrogen therapy results in downregulation of DNA repair genes. Next, we demonstrate that primary prostate cancers display a significant spectrum of AR transcriptional output, which correlates with expression of a set of DNA repair genes. Using RNA-seq and ChIP-seq, we define which of these DNA repair genes are both induced by androgen and represent direct AR targets. We establish that prostate cancer cells treated with ionizing radiation plus androgen demonstrate enhanced DNA repair and decreased DNA damage and furthermore that antiandrogen treatment causes increased DNA damage and decreased clonogenic survival. Finally, we demonstrate that antiandrogen treatment results in decreased classical nonhomologous end-joining. SIGNIFICANCE: We demonstrate that the AR regulates a network of DNA repair genes, providing a potential mechanism by which androgen deprivation synergizes with radiotherapy for prostate cancer.


Subject(s)
DNA Repair , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Androgen Antagonists/therapeutic use , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Cell Line, Tumor , DNA Damage/radiation effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Male , Metribolone/therapeutic use , Mice , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Radiation, Ionizing , Signal Transduction/genetics , Xenograft Model Antitumor Assays
17.
Nat Genet ; 44(6): 613-4, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22641202

ABSTRACT

The clinical spectrum of prostate cancer ranges from curable, local disease to widely metastatic, lethal cancer. Two new prostate cancer genome studies provide the first glimpse at both ends of this spectrum.


Subject(s)
Hepatocyte Nuclear Factor 3-alpha/genetics , Mediator Complex/genetics , Microtubule-Associated Proteins/genetics , Mutation , Prostatic Neoplasms/genetics , Humans , Male
18.
PLoS One ; 7(4): e34414, 2012.
Article in English | MEDLINE | ID: mdl-22509301

ABSTRACT

There is significant need to identify novel prostate cancer drug targets because current hormone therapies eventually fail, leading to a drug-resistant and fatal disease termed castration-resistant prostate cancer. To functionally identify genes that, when silenced, decrease prostate cancer cell proliferation or induce cell death in combination with antiandrogens, we employed an RNA interference-based short hairpin RNA barcode screen in LNCaP human prostate cancer cells. We identified and validated four candidate genes (AKT1, PSMC1, STRADA, and TTK) that impaired growth when silenced in androgen receptor positive prostate cancer cells and enhanced the antiproliferative effects of antiandrogens. Inhibition of AKT with a pharmacologic inhibitor also induced apoptosis when combined with antiandrogens, consistent with recent evidence for PI3K and AR pathway crosstalk in prostate cancer cells. Recovery of hairpins targeting a known prostate cancer pathway validates the utility of shRNA library screening in prostate cancer as a broad strategy to identify new candidate drug targets.


Subject(s)
Databases, Nucleic Acid , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Small Interfering/genetics , Androgen Antagonists/pharmacology , Anilides/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Genes, Neoplasm/genetics , Humans , Male , Molecular Targeted Therapy , Nitriles/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , RNA Interference , Recurrence , Reproducibility of Results , Tosyl Compounds/pharmacology
19.
PLoS One ; 6(3): e17449, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21394210

ABSTRACT

MYC and phosphoinositide 3-kinase (PI3K)-pathway deregulation are common in human prostate cancer. Through examination of 194 human prostate tumors, we observed statistically significant co-occurrence of MYC amplification and PI3K-pathway alteration, raising the possibility that these two lesions cooperate in prostate cancer progression. To investigate this, we generated bigenic mice in which both activated human AKT1 and human MYC are expressed in the prostate (MPAKT/Hi-MYC model). In contrast to mice expressing AKT1 alone (MPAKT model) or MYC alone (Hi-MYC model), the bigenic phenotype demonstrates accelerated progression of mouse prostate intraepithelial neoplasia (mPIN) to microinvasive disease with disruption of basement membrane, significant stromal remodeling and infiltration of macrophages, B- and T-lymphocytes, similar to inflammation observed in human prostate tumors. In contrast to the reversibility of mPIN lesions in young MPAKT mice after treatment with mTOR inhibitors, Hi-MYC and bigenic MPAKT/Hi-MYC mice were resistant. Additionally, older MPAKT mice showed reduced sensitivity to mTOR inhibition, suggesting that additional genetic events may dampen mTOR dependence. Since increased MYC expression is an early feature of many human prostate cancers, these data have implications for treatment of human prostate cancers with PI3K-pathway alterations using mTOR inhibitors.


Subject(s)
Precancerous Conditions/enzymology , Prostatic Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Disease Models, Animal , Disease Progression , Enzyme Activation/drug effects , Humans , Male , Mice , Mice, Transgenic , Neoplasm Invasiveness , Phenotype , Precancerous Conditions/pathology , Prostate/drug effects , Prostate/pathology , Prostatic Intraepithelial Neoplasia/enzymology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
20.
Cancer Cell ; 19(5): 575-86, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21575859

ABSTRACT

Prostate cancer is characterized by its dependence on androgen receptor (AR) and frequent activation of PI3K signaling. We find that AR transcriptional output is decreased in human and murine tumors with PTEN deletion and that PI3K pathway inhibition activates AR signaling by relieving feedback inhibition of HER kinases. Similarly, AR inhibition activates AKT signaling by reducing levels of the AKT phosphatase PHLPP. Thus, these two oncogenic pathways cross-regulate each other by reciprocal feedback. Inhibition of one activates the other, thereby maintaining tumor cell survival. However, combined pharmacologic inhibition of PI3K and AR signaling caused near-complete prostate cancer regressions in a Pten-deficient murine prostate cancer model and in human prostate cancer xenografts, indicating that both pathways coordinately support survival.


Subject(s)
PTEN Phosphohydrolase/deficiency , Phosphatidylinositol 3-Kinase/metabolism , Prostatic Neoplasms/enzymology , Receptors, Androgen/metabolism , Signal Transduction , Androgen Antagonists/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Mice, SCID , Mice, Transgenic , Nuclear Proteins/metabolism , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphoprotein Phosphatases/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/antagonists & inhibitors , Receptor, ErbB-3/metabolism , Receptors, Androgen/drug effects , Signal Transduction/drug effects , Time Factors , Transcription, Genetic , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL