Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Cancer Biol Ther ; 21(5): 424-431, 2020 05 03.
Article in English | MEDLINE | ID: mdl-32116106

ABSTRACT

Measurement of a tumor's overall genomic instability has gathered recent interest over the identification of specific genomic imbalances, as it may provide a more robust measure of tumor aggressiveness. Here we demonstrate the association of tumor genomic instability in the prediction of disease recurrence in patients with clinically localized clear cell renal cell carcinoma (ccRCC). Genomic copy number analysis was performed using SNP-based microarrays on tumors from 103 ccRCC patients. The number of copy number alterations (CNAs) for each tumor was calculated, and a genomic imbalance threshold (GIT) associated with high stage and high-grade disease was determined. Cox proportional hazards regression analyzes were performed to assess the effect of GIT on recurrence-free survival adjusting for known confounders. In the cohort, copy number losses in chromosome arms 3p, 14q, 6q, 9p, and 1p and gains of 5q and 7p/q were common. CNA burden significantly increased with increasing stage (p < .001) and grade (p < .001). The median CNA burden associated with patients presenting with advanced stage (IV) and high-grade (III/IV) tumors was ≥9, defining the GIT. On regression analysis, GIT was a superior predictor of recurrence (Hazard Ratio 4.44 [CI 1.36-14.48], p = .01) independent of stage, with similar results adjusting for grade. These findings were confirmed using an alternative measure of genomic instability, weighted Genomic Integrity Index. Our data support a key role for genomic instability in ccRCC progression. More importantly, we have identified a GIT (≥ 9 CNAs) that is a superior and independent predictor of disease recurrence in high-risk ccRCC patients.


Subject(s)
Adenocarcinoma, Clear Cell/mortality , DNA Copy Number Variations , Genomic Instability , Kidney Neoplasms/mortality , Neoplasm Recurrence, Local/mortality , Polymorphism, Single Nucleotide , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/pathology , Adult , Aged , Aged, 80 and over , DNA Copy Number Variations/genetics , Disease Progression , Female , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Prognosis , Survival Rate
4.
Neoplasia ; 20(1): 80-91, 2018 01.
Article in English | MEDLINE | ID: mdl-29216597

ABSTRACT

ONC201/TIC10 is a first-in-class small molecule inducer of TRAIL that causes early activation of the integrated stress response. Its promising safety profile and broad-spectrum efficacy in vitro have been confirmed in Phase I/II trials in several advanced malignancies. Binding and reporter assays have shown that ONC201 is a selective antagonist of the dopamine D2-like receptors, specifically, DRD2 and DRD3. We hypothesized that ONC201's interaction with DRD2 plays a role in ONC201's anticancer effects. Using cBioportal and quantitative reverse-transcription polymerase chain reaction analyses, we confirmed that DRD2 is expressed in different cancer cell types in a cell type-specific manner. On the other hand, DRD3 was generally not detectable. Overexpressing DRD2 in cells with low DRD2 levels increased ONC201-induced PARP cleavage, which was preceded and correlated with an increase in ONC201-induced CHOP mRNA expression. On the other hand, knocking out DRD2 using CRISPR/Cas9 in three cancer cell lines was not sufficient to abrogate ONC201's anticancer effects. Although ONC201's anticancer activity was not dependent on DRD2 expression in the cancer cell types tested, we assessed the cytotoxic potential of DRD2 blockade. Transient DRD2 knockdown in HCT116 cells activated the integrated stress response and reduced cell number. Pharmacological antagonism of DRD2 significantly reduced cell viability. Thus, we demonstrate in this study that disrupting dopamine receptor expression and activity can have cytotoxic effects that may at least be in part due to the activation of the integrated stress response. On the other hand, ONC201's anticancer activity goes beyond its ability to antagonize DRD2, potentially due to ONC201's ability to activate other pathways that are independent of DRD2. Nevertheless, blocking the dopamine D1-like receptor DRD5 via siRNA or the use of a pharmacological antagonist promoted ONC201-induced anticancer activity.


Subject(s)
Antineoplastic Agents/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neoplasms/metabolism , Receptors, Dopamine/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Humans , Imidazoles , Neoplasms/genetics , Pyridines , Pyrimidines , RNA, Small Interfering/genetics , Receptors, Dopamine/genetics , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/genetics , Receptors, Dopamine D3/metabolism , Receptors, Dopamine D5/genetics , Receptors, Dopamine D5/metabolism
5.
Sci Rep ; 7(1): 9711, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28851987

ABSTRACT

Nucleotide metabolism in cancer cells can influence malignant behavior and intrinsic resistance to therapy. Here we describe p53-dependent control of the rate-limiting enzyme in the pyrimidine catabolic pathway, dihydropyrimidine dehydrogenase (DPYD) and its effect on pharmacokinetics of and response to 5-fluorouracil (5-FU). Using in silico/chromatin-immunoprecipitation (ChIP) analysis we identify a conserved p53 DNA-binding site (p53BS) downstream of the DPYD gene with increased p53 occupancy following 5-FU treatment of cells. Consequently, decrease in Histone H3K9AC and increase in H3K27me3 marks at the DPYD promoter are observed concomitantly with reduced expression of DPYD mRNA and protein in a p53-dependent manner. Mechanistic studies reveal inhibition of DPYD expression by p53 is augmented following thymidylate synthase (TS) inhibition and DPYD repression by p53 is dependent on DNA-dependent protein kinase (DNA-PK) and Ataxia telangiectasia mutated (ATM) signaling. In-vivo, liver specific Tp53 loss increases the conversion of 5-FU to 5-FUH2 in plasma and elicits a diminished 5-FU therapeutic response in a syngeneic colorectal tumor model consistent with increased DPYD-activity. Our data suggest that p53 plays an important role in controlling pyrimidine catabolism through repression of DPYD expression, following metabolic stress imposed by nucleotide imbalance. These findings have implications for the toxicity and efficacy of the cancer therapeutic 5-FU.


Subject(s)
Dihydrouracil Dehydrogenase (NADP)/genetics , Gene Expression Regulation, Enzymologic , Pyrimidines/metabolism , Thymidylate Synthase/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, Liquid , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Humans , Male , Mice , Mice, Knockout , Models, Biological , Nuclear Proteins/metabolism , Polymorphism, Genetic , Protein Binding , Signal Transduction/drug effects , Tandem Mass Spectrometry , Tumor Suppressor Protein p53/genetics
7.
Curr Oncol Rep ; 17(12): 58, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26472299

ABSTRACT

Systemic therapy for urothelial carcinoma (UC) of the bladder has largely revolved around cytotoxic chemotherapy regimens. However, several recent clinical trials have explored the roles of targeted therapies which specifically inhibit signal transduction pathways. Simultaneously, a rationale for such therapies has come to the forefront of management of this disease because an overabundance of signaling pathways are genetically deranged as a result of point mutation or copy number alteration (CNA) as identified by several recent next generation sequencing (NGS) studies. Importantly, these derangements are found in all stages of disease, and therefore targeted therapies hold promise as a next step in the evolution of the medical management of both localized and metastatic UCC. We review the rationale for and progress in studying inhibition of signal transduction as a means of treatment of UCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Molecular Targeted Therapy/methods , Signal Transduction/drug effects , Urinary Bladder Neoplasms/drug therapy , Carcinoma, Transitional Cell/genetics , Clinical Trials as Topic , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Urinary Bladder Neoplasms/genetics
8.
J Endourol ; 27(6): 732-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23297710

ABSTRACT

BACKGROUND AND PURPOSE: Port-site metastasis (PSM) is a rare complication of laparoscopic intervention in urologic malignancies. Of the greater than 50 reported cases of PSM in the urologic oncology literature, only 9 have occurred after surgery for renal-cell carcinoma (RCC). We report a 10th instance of RCC metastasis-in this case to the camera-port site after robot-assisted partial nephrectomy (RAPN). To our knowledge, this case is the first reported PSM of RCC after RAPN. PATIENT AND METHODS: A 68-year-old man underwent an uncomplicated right RAPN for a 4-cm right renal mass (stage T1aN0M0). Five months later, he was found to have metastatic disease with an isolated peritoneal recurrence at the camera-port site. Biopsy of the lesion confirmed RCC, and the lesion was surgically resected. A comprehensive MEDLINE search for all published studies of port-site recurrences after laparoscopic renal surgery for RCC was performed. RESULTS: Nine cases of PSM after successful laparoscopic radical or partial nephrectomy for locally confined RCC have been reported. Proposed etiologic factors for port-site recurrence include biologic aggressiveness of the tumor, patient immunosuppression, local wound factors, and technique-related factors. We report an unusual case of PSM to a camera port that was not used for specimen manipulation or extraction. CONCLUSION: PSM after laparoscopic renal surgery for RCC is a rare occurrence. Our case, in which PSM occurred without specimen bag rupture or extraction through the port in question, highlights the importance of local and systemic factors in contributing to PSM occurrence. We also demonstrate that when PSM is the only site of disease recurrence, it can be successfully managed with minimally invasive surgical resection.


Subject(s)
Carcinoma, Renal Cell/secondary , Carcinoma, Renal Cell/surgery , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Neoplasm Seeding , Nephrectomy/adverse effects , Nephrectomy/methods , Robotics , Aged , Humans , Male
9.
J Sex Med ; 9(4): 1212-5, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22024210

ABSTRACT

INTRODUCTION: Inflatable penile prosthetic implants are a reliable treatment for erectile dysfunction. Mechanical failures now are the most common reason for revision of this type of device, and autoinflation is a common cause for device revision. There are currently no published surgical treatments for this malfunction. AIM: To describe a simple outpatient surgical revision for an automatically inflating device using laparascopic dissection. MAIN OUTCOME MEASURES: Complete deflation of penile prosthesis on follow-up visit, intraoperative and postsurgical complications, and length of procedure. METHODS: We performed a retrospective review of patients treated for inflatable penile prosthesis autoinflation with laparascopic capsulotomy to release constricting connective tissue rind surrounding the device reservoir at a single institution. We collected information about etiology of impotence, surgical procedures relating to implant and revision of prosthetic devices, and follow-up evaluations. RESULTS: Four patients underwent laparascopic capsulotomy to treat autoinflation. Mean operative time was 45 minutes, and no adverse surgical or perioperative outcomes occurred. All four patients had deflated corporal cylinders at the time of follow-up evaluation. CONCLUSIONS: Laparascopic capsulotomy is an easy and reliable method of treating inflatable penile prosthesis autoinflation that can be performed in the outpatient setting.


Subject(s)
Laparoscopy/methods , Penile Implantation/methods , Penile Prosthesis , Prosthesis Failure , Follow-Up Studies , Humans , Male , Patient Satisfaction , Postoperative Complications/surgery , Prosthesis Design , Reoperation , Retrospective Studies
11.
Semin Intervent Radiol ; 28(4): 361-6, 2011 Dec.
Article in English | MEDLINE | ID: mdl-23204634

ABSTRACT

Thermoablation (TA) has become an increasingly popular treatment for small renal masses (SRMs). Although long-term outcomes are not currently reported, TA may have a role in being an alternative to radical or partial nephrectomy. This review gives a broad overview of TA and discusses current controversies in the field.

12.
Clin Cancer Res ; 14(5): 1393-6, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18316560

ABSTRACT

PURPOSE: Carcinoids are neuroendocrine tumors and most frequently occur within tissues derived from the embryonic gut. These tumors can occur in any organ site but are rare in the testis. The cell type giving rise to testicular carcinoid is unknown. We hypothesized that testicular carcinoid may have a germ cell origin. EXPERIMENTAL DESIGN: We describe our analysis of protein and genetic markers of germ cell neoplasia, using immunohistochemistry and fluorescence in situ hybridization, in four testicular carcinoid tumors. RESULTS: All four cases of testicular carcinoid tumor arose in a background of mature teratoma. Isochromosome 12p was identified in carcinoid tumor cells in all four samples. 12p overrepresentation was also observed in three cases. Isochromosome 12p and 12p overrepresentation were present in cells of coexisting mature teratoma in three cases. Carcinoid tumors showed strong immunoreactivity for synaptophysin and chromogranin, but no immunoreactivity for OCT4, CD30, c-kit, TTF-1, and CDX2. Membranous and cytoplasmic staining for beta-catenin was detected in three cases. CONCLUSION: Our findings suggest that testicular carcinoid represents a phenotypic expression of testicular teratoma and is of germ cell origin.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoid Tumor/pathology , Testicular Neoplasms/pathology , Adult , Carcinoid Tumor/genetics , Carcinoid Tumor/metabolism , Chromogranins/metabolism , Chromosomes, Human, Pair 12/genetics , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Male , Synaptophysin/metabolism , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , beta Catenin/metabolism
13.
Mod Pathol ; 21(3): 355-62, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18192966

ABSTRACT

Small-cell carcinoma of the urinary bladder (SCBC) is a rare tumor, which shows a common clonal origin with urothelial carcinoma. It bears a high metastatic potential, even when discovered in a localized state. Identifying the molecular underpinnings of this disease may elucidate useful clinical information regarding prevention, diagnosis, prognosis, treatment, and surveillance. As DNA methylation is widely recognized as having a pivotal role in the process of carcinogenesis, we analyzed the DNA methylation status of four frequently hypermethylated tumor suppressors in small-cell and transitional-cell carcinoma (TCC) arising concomitantly in 13 patients. Fourteen cases of pure TCC were also included in the analysis. We identified frequent methylation of RASSF1 and MGMT and infrequent methylation of MLH1 and DAPK1 in cases of concomitant TCC and SCBC. Similar rates of methylation were found in pure and concomitant histopathologies, with the exception of MGMT, which was much less frequently methylated in pure TCC. These findings suggest that SCBC and TCC have common origins, establish DNA methylation of some tumor suppressors as frequent occurrences in both histopathologies, and suggest that MGMT methylation may be an SCBC-specific epimutation.


Subject(s)
Carcinoma, Small Cell/genetics , Carcinoma, Transitional Cell/genetics , CpG Islands/genetics , DNA Methylation , Genes, Tumor Suppressor , Urinary Bladder Neoplasms/genetics , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Carcinoma, Small Cell/pathology , Carcinoma, Transitional Cell/pathology , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Death-Associated Protein Kinases , Humans , MutL Protein Homolog 1 , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , Urinary Bladder Neoplasms/pathology
14.
Cancer Res ; 66(24): 11954-66, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17178894

ABSTRACT

The development of targeted therapies for antiestrogen-resistant breast cancer requires a detailed understanding of its molecular characteristics. To further elucidate the molecular events underlying acquired resistance to the antiestrogens tamoxifen and fulvestrant, we established drug-resistant sublines from a single colony of hormone-dependent breast cancer MCF7 cells. These model systems allowed us to examine the cellular and molecular changes induced by antiestrogens in the context of a uniform clonal background. Global changes in both basal and estrogen-induced gene expression profiles were determined in hormone-sensitive and hormonal-resistant sublines using Affymetrix Human Genome U133 Plus 2.0 Arrays. Changes in DNA methylation were assessed by differential methylation hybridization, a high-throughput promoter CpG island microarray analysis. By comparative studies, we found distinct gene expression and promoter DNA methylation profiles associated with acquired resistance to fulvestrant versus tamoxifen. Fulvestrant resistance was characterized by pronounced up-regulation of multiple growth-stimulatory pathways, resulting in estrogen receptor alpha (ERalpha)-independent, autocrine-regulated proliferation. Conversely, acquired resistance to tamoxifen correlated with maintenance of the ERalpha-positive phenotype, although receptor-mediated gene regulation was altered. Activation of growth-promoting genes, due to promoter hypomethylation, was more frequently observed in antiestrogen-resistant cells compared with gene inactivation by promoter hypermethylation, revealing an unexpected insight into the molecular changes associated with endocrine resistance. In summary, this study provides an in-depth understanding of the molecular changes specific to acquired resistance to clinically important antiestrogens. Such knowledge of resistance-associated mechanisms could allow for identification of therapy targets and strategies for resensitization to these well-established antihormonal agents.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , Estradiol/analogs & derivatives , Estrogen Receptor Modulators/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Tamoxifen/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Culture Media , Drug Resistance, Neoplasm , Estradiol/pharmacology , Fulvestrant , Humans , Oligonucleotide Array Sequence Analysis
15.
Cancer Res ; 66(11): 5582-91, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16740693

ABSTRACT

Histone modifications and DNA methylation are epigenetic phenomena that play a critical role in many neoplastic processes, including silencing of tumor suppressor genes. One such histone modification, particularly at H3 and H4, is methylation at specific lysine (K) residues. Whereas histone methylation of H3-K9 has been linked to DNA methylation and aberrant gene silencing in cancer cells, no such studies of H3-K27 have been reported. Here, we generated a stable cell line overexpressing a dominant-negative point mutant, H3-K27R, to examine the role of that specific lysine in ovarian cancer. Expression of this construct resulted in loss of methylation at H3-K27, global reduction of DNA methylation, and increased expression of tumor suppressor genes. One of the affected genes, RASSF1, was shown to be a direct target of H3-K27 methylation-mediated silencing. By increasing DNA-platinum adduct formation, indicating increased access of the drug to target DNA sequences, removal of H3-K27 methylation resensitized drug-resistant ovarian cancer cells to the chemotherapeutic agent cisplatin. This increased platinum-DNA access was likely due to relaxation of condensed chromatin. Our results show that overexpression of mutant H3-K27 in mammalian cells represents a novel tool for studying epigenetic mechanisms and the Histone Code Hypothesis in human cancer. Such findings show the significance of H3-K27 methylation as a promising target for epigenetic-based cancer therapies.


Subject(s)
Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic/physiology , Genes, Tumor Suppressor , Histones/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Point Mutation , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chromatin/metabolism , CpG Islands , DNA Methylation , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Gene Silencing , Humans , Lysine/genetics , Tumor Suppressor Proteins/genetics
16.
Thyroid ; 15(6): 551-61, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16029121

ABSTRACT

The beta-catenin pathway has been conclusively demonstrated to regulate differentiation and patterning in multiple model systems. In thyroid cancer, alterations are often seen in proteins that regulate beta-catenin, including those of the RAS, PI3K/AKT, and peroxisome proliferation activated receptor-gamma (PPARgamma) pathways, and evidence from the literature suggests that beta-catenin may play a direct role in the dedifferentiation commonly observed in late-stage disease. RET/PTC rearrangements are frequent in thyroid cancer and appear to be exclusive from mutational events in RAS and BRAF. Activation of AKT by phosphatidylinositide-3 kinase (PI3K), a RAS effector, results in GSK3beta phosphorylation and deactivation and subsequent beta-catenin upregulation in thyroid cancer. Activating mutations in beta-catenin, which have been demonstrated in late-stage thyroid tumors, correlate with beta-catenin nuclear localization and poor prognosis. We hypothesize that activation of the RAS, PI3K/AKT, and PPARgamma pathways ultimately impinges upon beta-catenin. We further propose that if mutations in BRAF, RAS, and RET/PTC rearrangements are mutually exclusive in certain thyroid tumors or tumor types, as has already been shown for papillary thyroid cancer, then these interconnected pathways may cooperate in the initiation and promotion of the disease. We believe that clinical benefit for thyroid cancer patients could be derived from disrupting the middle or distal pathway effectors of these pathways, such as AKT or beta-catenin.


Subject(s)
Signal Transduction , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , beta Catenin/metabolism , Genetic Therapy , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...