Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Pathol Int ; 72(3): 187-192, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35102630

ABSTRACT

NTRK fusions represent a new biomarker-defined population that can be treated with TRK inhibitors. Although rare, NTRK fusions are detected across a wide range of solid tumors. Previous reports suggest that NTRK fusions are limited to the secretory subtype of breast cancer. Here we examined NTRK fusions in a large real world next-generation sequencing (NGS) dataset and confirmed secretory versus non-secretory status using H&E images. Of 23 NTRK fusion-positive cases, 11 were classified as secretory, 11 as non-secretory, and one as mixed status. The secretory subtype trended younger, was predominantly estrogen receptor (ER)-, had lower tumor mutational burden, and exhibited lower levels of genomic loss of heterozygosity. The non-secretory subtype was enriched for TP53 mutations. The secretory subtype was enriched for ETV6-NTRK3 fusions in 7 of 11 cases, and the non-secretory subtype had NTRK1 fusions in 7 of 11 cases, each with a different fusion partner. Our data suggests NTRK fusions are present in both secretory and non-secretory subtypes, and that comprehensive genomic profiling should be considered across all clinically advanced breast cancers to identify patients that could receive benefit from TRK inhibitors.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/diagnosis , Receptor, trkA/genetics , Aged , Breast Neoplasms/diagnosis , Carcinoma/genetics , Female , Gene Fusion/drug effects , Gene Fusion/genetics , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/statistics & numerical data , Humans , Immunohistochemistry/methods , Immunohistochemistry/statistics & numerical data , Middle Aged , Receptor, trkA/adverse effects , Receptor, trkC/genetics
2.
Int J Hematol ; 115(1): 43-53, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34546543

ABSTRACT

All-trans retinoic acid (ATRA)-based therapy for acute promyelocytic leukemia (APL), a subtype of acute myeloid leukemia (AML), is the most successful example of differentiation therapy. Although ATRA can induce differentiation in some non-APL AML cell lines and primary blasts, clinical results of adding ATRA to standard therapy in non-APL AML patients have been inconsistent, probably due to use of different regimens and lack of diagnostic tools for identifying which patients may be sensitive to ATRA. In this study, we exposed primary blasts obtained from non-APL AML patients to ATRA to test for differentiation potential in vitro. We observed increased expression of differentiation markers, indicating a response to ATRA, in four out of fifteen primary AML samples. Three samples in which CD11b increased in response to ATRA had an inversion of chromosome 16 as well as the CBFB-MYH11 fusion gene, and the fourth sample was from a patient with KMT2A-rearranged, therapy-related AML. In conclusion, we identified a subgroup of non-APL AML patients with inv(16) and CBFB-MYH11 as the most sensitive to ATRA-mediated differentiation in vitro, and our results can help identify patients who may benefit from ATRA treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Blast Crisis/genetics , Blast Crisis/pathology , Chromosome Inversion/genetics , Chromosomes, Human, Pair 16/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Tretinoin/pharmacology , Tretinoin/therapeutic use , CD11b Antigen/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Core Binding Factor beta Subunit/genetics , Gene Fusion/drug effects , Gene Rearrangement/genetics , Histone-Lysine N-Methyltransferase/genetics , Humans , Myeloid-Lymphoid Leukemia Protein/genetics , Myosin Heavy Chains/genetics
3.
Clin Cancer Res ; 27(11): 3154-3166, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33824166

ABSTRACT

PURPOSE: Oncogenic fusions involving the neuregulin 1 (NRG1) gene are found in approximately 0.2% of cancers of diverse histologies. The resulting chimeric NRG1 proteins bind predominantly to HER3, leading to HER3-HER2 dimerization and activation of downstream growth and survival pathways. HER3 is, therefore, a rational target for therapy in NRG1 fusion-driven cancers. EXPERIMENTAL DESIGN: We developed novel patient-derived and isogenic models of NRG1-rearranged cancers and examined the effect of the anti-HER3 antibody, seribantumab, on growth and activation of signaling networks in vitro and in vivo. RESULTS: Seribantumab inhibited NRG1-stimulated growth of MCF-7 cells and growth of patient-derived breast (MDA-MB-175-VII, DOC4-NRG1 fusion) and lung (LUAD-0061AS3, SLC3A2-NRG1 fusion) cancer cells harboring NRG1 fusions or NRG1 amplification (HCC-95). In addition, seribantumab inhibited growth of isogenic HBEC cells expressing a CD74-NRG1 fusion (HBECp53-CD74-NRG1) and induced apoptosis in MDA-MB-175-VII and LUAD-0061AS3 cells. Induction of proapoptotic proteins and reduced expression of the cell-cycle regulator, cyclin D1, were observed in seribantumab-treated cells. Treatment of MDA-MB-175-VII, LUAD-0061AS3, and HBECp53-CD74-NRG1 cells with seribantumab reduced phosphorylation of EGFR, HER2, HER3, HER4, and known downstream signaling molecules, such as AKT and ERK1/2. Significantly, administration of seribantumab to mice bearing LUAD-0061AS3 patient-derived xenograft (PDX) and OV-10-0050 (ovarian cancer with CLU-NRG1 fusion) PDX tumors induced regression of tumors by 50%-100%. Afatinib was much less effective at blocking tumor growth. CONCLUSIONS: Seribantumab treatment blocked activation of the four ERBB family members and of downstream signaling, leading to inhibition of NRG1 fusion-dependent tumorigenesis in vitro and in vivo in breast, lung, and ovarian patient-derived cancer models.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression/drug effects , Gene Fusion/drug effects , Gene Fusion/genetics , Neoplasms/genetics , Neoplasms/pathology , Neuregulin-1/genetics , Neuregulin-1/metabolism , Receptor, ErbB-3/immunology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells , Mice , Protein Binding , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Xenograft Model Antitumor Assays
4.
J Cancer Res Clin Oncol ; 147(2): 323-337, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33387037

ABSTRACT

The incidence of papillary thyroid cancer (PTC), the major type of thyroid cancer, is increasing rapidly around the world, and its pathogenesis is still unclear. There is poor prognosis for PTC involved in rapidly progressive tumors and resistance to radioiodine therapy. Kinase gene fusions have been discovered to be present in a wide variety of malignant tumors, and an increasing number of novel types have been detected in PTC, especially progressive tumors. As a tumor-driving event, kinase fusions are constitutively activated or overexpress their kinase function, conferring oncogenic potential, and their frequency is second only to BRAFV600E mutation in PTC. Diverse forms of kinase fusions have been observed and are associated with specific pathological features of PTC (usually at an advanced stage), and clinical trials of therapeutic strategies targeting kinase gene fusions are feasible for radioiodine-resistant PTC. This review summarizes the roles of kinase gene fusions in PTC and the value of clinical therapy of targeting fusions in progressive or refractory PTC, and discusses the future perspectives and challenges related to kinase gene fusions in PTC patients.


Subject(s)
Gene Fusion , Protein Kinases/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Anaplastic Lymphoma Kinase/genetics , Gene Fusion/drug effects , Gene Fusion/physiology , Humans , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-ret/genetics , Receptor, trkA/genetics , Thyroid Cancer, Papillary/drug therapy , Thyroid Neoplasms/drug therapy
5.
Medicine (Baltimore) ; 98(3): e14120, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30653139

ABSTRACT

RATIONALE: Lung cancer is a series of gene-driven disease. EGFR, ALK, and ROS1 are 3 major driver genes that play an important role in lung cancer development and precision management. Additionally, rare genetic alterations continue to be discovered and may become novel targets for therapy. The RET gene is one of such rare genetic alteration of non-small cell lung cancer (NSCLC). In this report, we present a RET-positive case that benefited from cabozantinib treatment. PATIENT CONCERN: A 50-year-old male patient was diagnosed with lung adenocarcinoma 2 years ago, at that time he received palliative surgery of pulmonary carcinoma and completed 4 cycles of chemotherapy with gemcitabine and cisplatin. Six months later, he was hospitalized in our cancer center due to the disease recurrence, presenting with pleural metastasis. DIAGNOSIS: Gene alteration was examined using the intraoperative specimen by PCR method, and KIF5B/RET gene fusion was detected. Therefore, the patient was diagnosed with late-stage lung adenocarcinoma with RET gene mutation. INTERVENTIONS: The patient received treatment with cabozantinib from June 2017. OUTCOMES: Cabozantinib was administered (140 mg orally, once daily) for approximate 9 months, and his disease achieved stable disease (SD). During that period, there were no severe adverse events (AE), except for a grade II rash (CTCAE 4.0). LESSONS: We found that the RET fusion gene is a novel driver molecular of lung adenocarcinoma in patients without common mutations in such genes as EGFR, ALK, and ROS1. This case report supports a rationale for the treatment of lung adenocarcinoma patients with a RET fusion and provides alternative treatment options for these types of NSCLC patients.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Anilides/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-ret/genetics , Pyridines/therapeutic use , Adenocarcinoma of Lung/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Gene Fusion/drug effects , Humans , Kinesins/genetics , Lung Neoplasms/genetics , Male , Middle Aged , Proto-Oncogene Proteins c-ret/drug effects
6.
Brain Tumor Pathol ; 36(1): 14-19, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30350109

ABSTRACT

The proto-oncogene tyrosine-protein kinase ROS1 (ROS1) is a tyrosine kinase that is closely related to anaplastic lymphoma kinase receptor (ALK). We describe a novel KLC1-ROS1 fusion identified in a case of pediatric low-grade glioma. This was detected by RNA sequencing and confirmed by reverse-transcription PCR and fluorescent in situ hybridization. Immunohistochemical staining for ROS1 was positive in the tumor cytoplasm. In vitro analysis demonstrated the oncogenic activity of this fusion, which was suppressed by the ALK/ROS1 inhibitor, crizotinib. Our case and others suggest that various ROS1 fusions might be present in a subset of pediatric gliomas, which could be targeted for therapy.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Gene Fusion , Glioma/genetics , Glioma/pathology , Microtubule-Associated Proteins/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Brain Neoplasms/drug therapy , Child, Preschool , Crizotinib/pharmacology , Female , Gene Fusion/drug effects , Glioma/drug therapy , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kinesins , Molecular Targeted Therapy , Neoplasm Staging , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
Medicine (Baltimore) ; 97(52): e13797, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30593165

ABSTRACT

RATIONALE: Although most of non-small cell lung cancer (NSCLC) patients with ROS1-fusions respond to crizotinb, acquired resistance eventually develop. The next-generations of ROS1 inhibitors have made some achievements, but the effects of immunotherapy have not been explored. PATIENT CONCERNS: A 44-year-old Chinese women presented with cough and dyspnea with a history of advanced lung adenocarcinoma. DIAGNOSIS: A PET/CT scan revealed primary tumors in bilateral lung lobes and multiple metastases in lymph nodes and bones. And ultrasound-guided left cervical lymph node biopsy revealed the pathological diagnosis was poor differentiated lung adenocarcinoma. INTERVENTIONS: The patients was started to be treated with 4 cycles of pemetrexed, carboplatin and bevacizumab, followed by one cycle of docetaxel, cisplatin and bevacizumab. As the ROS1-fusion was found by next generation sequencing, the patient received crizotinib treatment about 3 months. OUTCOMES: After 5 cycles of chemotherapy, CT scans revealed increased size of bilateral lobe nodules indicative of progressive disease (PD). Then the patient received treatment of crizotinib and his progression-free survival reached 3 months. Due to uncontrollable disease progression, the patient expired. LESSONS: The genetic profile of NSCLC patients might be altered in various therapeutic processes. Thus, repeated genetic testing might be important at each progression. Moreover, immunotherapy might be a powerful weapon to overcome the resistance to Tyrosine kinase inhibitors (TKIs) in future.


Subject(s)
Adenocarcinoma of Lung/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Crizotinib/therapeutic use , Drug Resistance, Neoplasm/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Adult , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Fatal Outcome , Female , Gene Fusion/drug effects , Gene Fusion/genetics , Humans , Mutation/drug effects , Mutation/genetics , Progression-Free Survival , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/drug effects , Treatment Failure , Tumor Burden
8.
Pac Symp Biocomput ; 23: 8-19, 2018.
Article in English | MEDLINE | ID: mdl-29218865

ABSTRACT

We characterize the transcriptional splicing landscape of a prostate cancer cell line treated with a previously identified synergistic drug combination. We use a combination of third generation long-read RNA sequencing technology and short-read RNAseq to create a high-fidelity map of expressed isoforms and fusions to quantify splicing events triggered by treatment. We find strong evidence for drug-induced, coherent splicing changes which disrupt the function of oncogenic proteins, and detect novel transcripts arising from previously unreported fusion events.


Subject(s)
Alternative Splicing/drug effects , Alternative Splicing/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Antineoplastic Combined Chemotherapy Protocols , Cell Line, Tumor , Computational Biology , Gene Expression Profiling , Gene Fusion/drug effects , Humans , Male , Mefloquine/administration & dosage , RNA Splicing/drug effects , RNA Splicing/genetics , RNA, Neoplasm/genetics , Sequence Analysis, RNA , Tamoxifen/administration & dosage
9.
Prostate ; 77(3): 282-290, 2017 02.
Article in English | MEDLINE | ID: mdl-27753122

ABSTRACT

BACKGROUND: Calcium channel blockers (CCBs) may affect prostate cancer (PCa) growth by various mechanisms including those related to androgens. The fusion of the androgen-regulated gene TMPRSS2 and the oncogene ERG (TMPRSS2:ERG or T2E) is common in PCa, and prostate tumors that harbor the gene fusion are believed to represent a distinct disease subtype. We studied the association of CCB use with the risk of PCa, and molecular subtypes of PCa defined by T2E status. METHODS: Participants were residents of King County, Washington, recruited for population-based case-control studies (1993-1996 or 2002-2005). Tumor T2E status was determined by fluorescence in situ hybridization using tumor tissue specimens from radical prostatectomy. Detailed information on use of CCBs and other variables was obtained through in-person interviews. Binomial and polytomous logistic regression were used to generate odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS: The study included 1,747 PCa patients and 1,635 age-matched controls. A subset of 563 patients treated with radical prostatectomy had T2E status determined, of which 295 were T2E positive (52%). Use of CCBs (ever vs. never) was not associated with overall PCa risk. However, among European-American men, users had a reduced risk of higher-grade PCa (Gleason scores ≥7: adjusted OR = 0.64; 95% CI: 0.44-0.95). Further, use of CCBs was associated with a reduced risk of T2E positive PCa (adjusted OR = 0.38; 95% CI: 0.19-0.78), but was not associated with T2E negative PCa. CONCLUSIONS: This study found suggestive evidence that use of CCBs is associated with reduced relative risks for higher Gleason score and T2E positive PCa. Future studies of PCa etiology should consider etiologic heterogeneity as PCa subtypes may develop through different causal pathways. Prostate 77:282-290, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Calcium Channel Blockers/therapeutic use , Gene Fusion/genetics , Population Surveillance , Prostatic Neoplasms/genetics , Serine Endopeptidases/genetics , Aged , Calcium Channel Blockers/pharmacology , Case-Control Studies , Gene Fusion/drug effects , Humans , Male , Middle Aged , Population Surveillance/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Risk Factors , Serine Endopeptidases/metabolism , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism
10.
Zhongguo Fei Ai Za Zhi ; 17(10): 750-4, 2014 Oct 20.
Article in Chinese | MEDLINE | ID: mdl-25342042

ABSTRACT

Lung cancer is the leading cause of cancer-related mortality in the worldwide. The discovery of drive gene makes tumor treatment is no longer "one-size-fits-all". Targeted therapy to change the present situation of cancer drugs become "bullet" with eyes, the effect is visible and bring a revolution in the treatment of lung cancer. The diver gene and targeted therapy have became the new cedule of non-small cell lung cancer (NSCLC). Society of Clinical Oncology (ASCO) has showed 11 kinds of diver genes. Here, we review the functional and structural characteristics and the targeted therapy in the 11 kinds of driver gene mutations.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Genes, Neoplasm/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Molecular Targeted Therapy/methods , Carcinoma, Non-Small-Cell Lung/pathology , Gene Fusion/drug effects , Humans , Signal Transduction/drug effects
11.
Prostate ; 73(1): 101-12, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22674191

ABSTRACT

BACKGROUND: Although endocrine therapy has been used for decades, its influence on the expression of microRNAs (miRNAs) in clinical tissue specimens has not been analyzed. Moreover, the effects of the TMPRSS2:ERG fusion on the expression of miRNAs in hormone naïve and endocrine-treated prostate cancers are poorly understood. METHODS: We used clinical material from a neoadjuvant trial consisting of 28 men treated with goserelin (n = 8), bicalutamide (n = 9), or no treatment (n = 11) for 3 months prior to radical prostatectomy. Freshly frozen specimens were used for microarray analysis of 723 human miRNAs. Specific miRNA expression in cancer, benign epithelium and stromal tissue compartments was predicted with an in silico Bayesian modeling tool. RESULTS: The expression of 52, 44, and 34 miRNAs was affected >1.4-fold by the endocrine treatment in the cancer, non-malignant epithelium, and stromal compartments, respectively. Of the 52 miRNAs, only 10 were equally affected by the two treatment modalities in the cancer compartment. Twenty-six of the 52 genes (50%) showed AR binding sites in their proximity in either VCaP or LNCaP cell lines. Forty-seven miRNAs were differentially expressed in TMPRSS2:ERG fusion positive compared with fusion negative cases. Endocrine treatment reduced the differences between fusion positive and negative cases. CONCLUSIONS: Goserelin treatment and bicalutamide treatment mostly affected the expression of different miRNAs. The effect clearly varied in different tissue compartments. TMPRSS2:ERG fusion positive and negative cases showed differential expression of miRNAs, and the difference was diminished by androgen ablation.


Subject(s)
Adenocarcinoma/drug therapy , Anilides/therapeutic use , Antineoplastic Agents/therapeutic use , Goserelin/therapeutic use , MicroRNAs/genetics , Nitriles/therapeutic use , Prostatic Neoplasms/drug therapy , Tosyl Compounds/therapeutic use , Adenocarcinoma/metabolism , Computer Simulation , Gene Expression Regulation, Neoplastic/drug effects , Gene Fusion/drug effects , Humans , In Situ Hybridization, Fluorescence , Male , MicroRNAs/metabolism , Neoadjuvant Therapy , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Prostatectomy , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured
12.
Trends Endocrinol Metab ; 22(12): 474-80, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21889355

ABSTRACT

The androgen receptor (AR) is important for prostate cancer development and progression. Genome-wide mapping of AR binding sites in prostate cancer has found that the majority of AR binding sites are located within non-promoter regions. These distal AR binding regions regulate AR target genes (e.g. UBE2C) involved in prostate cancer growth through chromatin looping. In addition to long-distance gene regulation, looping has been shown to induce spatial proximity of two genes otherwise located far away along the genomic sequence and the formation of double-strand DNA breaks, resulting in aberrant gene fusions (e.g. TMPRSS2-ERG) that also contribute to prostate tumorigenesis. Elucidating the mechanisms of AR-driven chromatin looping will increase our understanding of prostate carcinogenesis and may lead to the identification of new therapeutic targets.


Subject(s)
Androgens/metabolism , Chromatin Assembly and Disassembly , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Androgens/adverse effects , Animals , Carcinogens/metabolism , Carcinogens/toxicity , Gene Fusion/drug effects , Humans , Male , Molecular Targeted Therapy , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Neoplasm Proteins/genetics , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Signal Transduction/drug effects
13.
PLoS One ; 6(6): e21319, 2011.
Article in English | MEDLINE | ID: mdl-21731703

ABSTRACT

Prostate specific membrane antigen (PSMA) is overexpressed in prostatic adenocarcinoma (CaP), and its expression is negatively regulated by androgen stimulation. However, it is still unclear which factors are involved in this downregulation. TMPRSS2-ERG fusion is the most common known gene rearrangement in prostate carcinoma. Androgen stimulation can increase expression of the TMPRSS2-ERG fusion in fusion positive prostate cancer cells. The purpose of this investigation is to determine whether PSMA expression can be regulated by the TMPRSS2-ERG gene fusion. We employed two PSMA positive cell lines: VCaP cells, which harbor TMPRSS2-ERG fusion, and LNCaP cells, which lack the fusion. After 24 hours of androgen treatment, TMPRSS2-ERG mRNA level was increased in VCaP cells. PSMA mRNA level was dramatically decreased in VCaP cells, while it only has moderate change in LNCaP cells. Treatment with the androgen antagonist flutamide partially restored PSMA expression in androgen-treated VCaP cells. Knocking down ERG by siRNA in VCaP cells enhances PSMA expression both in the presence and absence of synthetic androgen R1881. Overexpressing TMPRSS2-ERG fusions in LNCaP cells downregulated PSMA both in the presence or absence of R1881, while overexpressing wild type ERG did not. Using PSMA-based luciferase reporter assays, we found TMPRSS2-ERG fusion can inhibit PSMA activity at the transcriptional level. Our data indicated that downregulation of PSMA in androgen-treated VCaP cells appears partially mediated by TMPRSS2-ERG gene fusion.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Fusion , Oncogene Proteins, Fusion/genetics , Prostate-Specific Antigen/genetics , Base Sequence , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Fusion/drug effects , Gene Knockdown Techniques , Humans , Luciferases/metabolism , Male , Metribolone/pharmacology , Molecular Sequence Data , Prostate-Specific Antigen/antagonists & inhibitors , Prostate-Specific Antigen/metabolism , RNA, Small Interfering/metabolism
14.
Cancer Res ; 70(23): 9544-8, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20947519

ABSTRACT

Fusion genes play important roles in tumorigenesis. The identification of the high-frequency TMPRSS2 fusion with ERG and other ETS family genes in prostate cancer highlights the importance of fusion genes in solid tumor development and progression. However, the mechanisms leading to these fusions are unclear. We investigated whether androgen, through stimulating its receptor, could promote spatial genome reorganization and contribute to the generation of the TMPRSS2:ERG fusion. We show that treatment with androgen can induce the TMPRSS2:ERG fusion in both malignant and nonmalignant prostate epithelial cells. Although the fusion could be detected in malignant cells following 24-hour treatment, prolonged exposure to androgen was required to detect the fusion transcript in nonmalignant cells. We associated the fusion incidence with genetic factors, including androgen-induced gene proximity, androgen receptor exon1 CAG repeat length and expression of the PIWIL1 gene. This study demonstrates that fusions can be induced prior to malignant transformation and generation of the fusion is associated with both gene proximity and loss of the ability to prevent double-strand breaks.


Subject(s)
Dihydrotestosterone/pharmacology , Epithelial Cells/drug effects , Gene Fusion/drug effects , Oncogene Proteins, Fusion/genetics , Androgens/pharmacology , Argonaute Proteins , Cell Line, Transformed , Cell Line, Tumor , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Situ Hybridization, Fluorescence , Male , Prostate/cytology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...