Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Arch Pathol Lab Med ; 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38452805

ABSTRACT

CONTEXT.­: Mutant KRAS is the main oncogenic driver in pancreatic ductal adenocarcinomas (PDACs). However, the clinical and phenotypic implications of harboring different mutant KRAS alleles remain poorly understood. OBJECTIVE.­: To characterize the potential morphologic and clinical outcome differences in PDACs harboring distinct mutant KRAS alleles. DESIGN.­: Cohort 1 consisted of 127 primary conventional PDACs with no neoadjuvant therapy, excluding colloid/mucinous, adenosquamous, undifferentiated, and intraductal papillary mucinous neoplasm-associated carcinomas, for which an in-house 42-gene mutational panel had been performed. A morphologic classification system was devised wherein each tumor was assigned as conventional, papillary/large duct (P+LD, defined as neoplastic glands with papillary structure and/or with length ≥0.5 mm), or poorly differentiated (when the aforementioned component was 60% or more of the tumor). Cohort 2 was a cohort of 88 PDACs in The Cancer Genome Atlas, which were similarly analyzed. RESULTS.­: In both cohorts, there was significant enrichment of P+LD morphology in PDACs with KRAS G12V and G12R compared with G12D. In the entire combined cohort, Kaplan-Meier analyses showed longer overall survival (OS) with KRAS G12R as compared with G12D (median OS of 1255 versus 682 days, P = .03) and in patients whose PDACs displayed P+LD morphology as compared with conventional morphology (median OS of 1175 versus 684 days, P = .04). In the adjuvant-only subset, KRAS G12R had the longest OS compared with G12D, G12V, and other alleles (median OS unreached/undefined versus 1009, 1129, and 1222 days, respectively). CONCLUSIONS.­: PDACs with different mutant KRAS alleles are associated with distinct morphologies and clinical outcomes, with KRAS G12R allele associated with P+LD morphology and longer OS when compared with G12D using Kaplan-Meier studies.

2.
J Exp Clin Cancer Res ; 41(1): 41, 2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35086548

ABSTRACT

BACKGROUND: Cancer cells have an imbalance in oxidation-reduction (redox) homeostasis. Understanding the precise mechanisms and the impact of the altered redox microenvironment on the immunologic reaction to tumors is limited. METHODS: We isolated exosomes from ovarian cancer cells through ultracentrifuge and characterized by Western-blots and Nanoparticle Tracking Analysis. 2D, 3D-coculture tumor model, and 3D live cell imaging were used to study the interactions between tumor cells, macrophages and CD3 T cells in vitro. The role of exosomal miR-155-5p in tumor growth was evaluated in xenograft nude mice models and immune-competent mice models. Flow cytometry and flow sorting were used to determine the expression levels of miR-155-5p and PD-L1 in ascites and splenic macrophages, and the percentages of CD3 T cells subpopulations. RESULTS: The elevation of reactive oxygen species (ROS) greatly downregulated exosomal miR-155-5p expression in tumor cells. Neutralization of ROS with N-acetyl-L-cysteine (NAC) increased the levels of miR-155-5p in tumor exosomes that were taken up by macrophages, leading to reduction of macrophage migration and tumor spheroid infiltration. We further found that programmed death ligand 1 (PD-L1) is a functional target of miR-155-5p. Co-culture of macrophages pre-treated with NAC-derived tumor exosomes or exosomal miR-155-5p with T-lymphocytes leading to an increased percentage of CD8+ T-lymphocyte and a decreased CD3+ T cell apoptosis through PD-L1 downregulation. Tumor growth in nude mice was delayed by treatment with NAC-derived tumor exosomes. Delivery of tumor exo-miR-155-5p in immune-intact mice suppressed ovarian cancer progression and macrophage infiltration, and activated CD8+ T cell function. It is of note that exo-miR-155-5p inhibited tumor growth more potently than the PD-L1 antibody, suggesting that in addition to PD-L1, other pathways may also be targeted by this approach. CONCLUSIONS: Our findings demonstrate a novel mechanism, ROS-induced down-regulation of miR-155-5p, by which tumors modulate the microenvironment that favors tumor growth. Understanding of the negative impact of ROS on the tumor immune response will improve current therapeutic strategies. Targeting miR-155-5p can be an alternative approach to prevent formation of an immunosuppressive TME through downregulation of PD-L1 and other immunosuppressive factors.


Subject(s)
B7-H1 Antigen/metabolism , Exosomes/immunology , Immunity/immunology , Macrophages/metabolism , MicroRNAs/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Reactive Oxygen Species
3.
Acta Med Acad ; 50(1): 4-12, 2021 Apr.
Article in English | MEDLINE | ID: mdl-34075760

ABSTRACT

OBJECTIVE: Mutations in the EIF1AX gene have been recently detected in a small percentage of benign and malignant thyroid lesions. We sought to investigate the prevalence and clinical significance of EIF1AX mutations and co-mutations in cytologically indeterminate thyroid nodules at our institution. MATERIALS AND METHODS: A 5-year retrospective analysis was performed on thyroid nodules with a cytologic diagnosis of Bethesda category III or IV, which had undergone testing by our in-house next generation sequencing panel. Surgically resected nodules with EIF1AX mutations were identified, and mutation type and presence of co-mutations were correlated with histopathologic diagnosis. RESULTS: 41/904 (4.5%) cases overall and 26/229 (11.4%) surgically resected nodules harbored an EIF1AX mutation. The most common histologic diagnoses were follicular thyroid carcinoma and follicular variant of papillary thyroid carcinoma. 11/26 (42.3%) of nodules had isolated EIF1AX mutation. Comutations were found in RAS (12/26; 46.2%), TERT (5/26; 19.2%) and TP53 (2/26; 7.7%). EIF1AX mutation alone conferred a 36.4% risk of malignancy (ROM) and 54.5% ROM or noninvasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP), while the ROM was significantly higher in nodules with concurrent RAS (71.4%), TERT, TP53 and RAS+TERT (100%) mutations. CONCLUSION: EIF1AX mutations occur in benign and malignant follicular thyroid neoplasms. In our cohort, the majority of mutations occurred at the splice acceptor site between exons 5 and 6. Importantly, the coexistence of EIF1AX mutations with other driver pathogenic mutations in RAS, TERT and TP53 conferred a 100% ROM or NIFTP, indicating that such nodules require surgical removal.


Subject(s)
Adenocarcinoma, Follicular , Eukaryotic Initiation Factor-1 , Thyroid Neoplasms , Thyroid Nodule , Eukaryotic Initiation Factor-1/genetics , Humans , Mutation , Retrospective Studies , Thyroid Neoplasms/genetics , Thyroid Nodule/genetics
5.
Oncogene ; 39(49): 7181-7195, 2020 12.
Article in English | MEDLINE | ID: mdl-33037411

ABSTRACT

The development of resistance to EGFR Tyrosine kinase inhibitors (TKIs) in NSCLC with activating EGFR mutations is a critical limitation of this therapy. In addition to genetic alterations such as EGFR secondary mutation causing EGFR-TKI resistance, compensatory activation of signaling pathways without interruption of genome integrity remains to be defined. In this study, we identified S6K1/MDM2 signaling axis as a novel bypass mechanism for the development of EGFR-TKI resistance. The observation of S6K1 as a candidate mechanism for resistance to EGFR TKI therapy was investigated by interrogation of public databases and a clinical cohort to establish S6K1 expression as a prognostic/predictive biomarker. The role of S6K1 in TKI resistance was determined in in vitro gain-and-loss of function studies and confirmed in subcutaneous and orthotopic mouse lung cancer models. Blockade of S6K1 by a specific inhibitor PF-4708671 synergistically enhanced the efficacy of TKI without showing toxicity. The mechanistic study showed the inhibition of EGFR caused nuclear translocation of S6K1 for binding with MDM2 in resistant cells. MDM2 is a downstream effector of S6K1-mediated TKI resistance. Taken together, we present evidence for the reversal of resistance to EGFR TKI by the addition of small molecule S6K1/MDM2 antagonists that could have clinical benefit.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Adult , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , ErbB Receptors/antagonists & inhibitors , Humans , Male , Mice , Mutation , Prognosis , Proto-Oncogene Proteins c-mdm2/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
6.
Arch Pathol Lab Med ; 144(11): 1303-1310, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32649229

ABSTRACT

CONTEXT.­: We implemented multiple nucleic acid amplification test platforms because of the limited availability of test kits for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) during the early stages of the pandemic. Interpretation of results generated by different platforms and prioritization for testing algorithms required cross-comparison. OBJECTIVE.­: To compare the analytical sensitivity of 3 commercial SARS-CoV-2 molecular assays, selected samples were studied in parallel with Cobas SARS-CoV-2 test, NxTAG CoV Extended Panel, and ID NOW COVID-19 assays. DESIGN.­: A total of 8043 SARS-CoV-2 tests performed from March 22 to April 19, 2020, were included in this study. For all 1794 positive specimens detected by the cobas SARS-CoV-2 assay, the cycle threshold (Ct) values were manually tracked and plotted to demonstrate the distribution of sample viral levels. Additionally, 50 and 63 low-positive specimens (Ct values >32) as well as 50 and 61 consecutive positive specimens by the cobas assay were tested with NxTAG and ID NOW, respectively, to estimate their relative sensitivities. RESULTS.­: The Ct values of cobas SARS-CoV-2-positive samples were evenly distributed throughout ranges of 13.32 to 39.50 (mean, 25.06) and 13.60 to 42.49 (mean, 26.45) for ORF1 and E gene targets, respectively. NxTAG reliably detected only specimens with E gene Ct values lower than 33, and is estimated to detect 89.4% of positive specimens detected by cobas assay. ID NOW had performance variation independent of Ct value and is estimated to detect 83.5% of cobas positives. CONCLUSIONS.­: Clinical specimens exhibit a wide range of viral burden, with a significant portion at low levels. Analytical sensitivity of testing platforms is critical for reliable detection of SARS-CoV-2 and uniform care to patients.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/diagnosis , Molecular Diagnostic Techniques/methods , Nucleic Acid Amplification Techniques/methods , Pneumonia, Viral/diagnosis , Severe Acute Respiratory Syndrome/diagnosis , Severe acute respiratory syndrome-related coronavirus/genetics , Adult , Betacoronavirus/isolation & purification , Betacoronavirus/physiology , COVID-19 , Clinical Laboratory Techniques/methods , Coronavirus Infections/virology , Diagnosis, Differential , Early Diagnosis , Female , Humans , Male , Middle Aged , Nasopharynx/pathology , Nasopharynx/virology , Pandemics , Pneumonia, Viral/virology , Severe acute respiratory syndrome-related coronavirus/isolation & purification , Severe acute respiratory syndrome-related coronavirus/physiology , SARS-CoV-2 , Sensitivity and Specificity , Severe Acute Respiratory Syndrome/virology , Specimen Handling/instrumentation , Specimen Handling/methods
7.
J Neurol Neurosurg Psychiatry ; 91(8): 846-848, 2020 08.
Article in English | MEDLINE | ID: mdl-32354770

ABSTRACT

BACKGROUND: Emergence of the novel corona virus (severe acute respiratory syndrome (SARS)-CoV-2) in December 2019 has led to the COVID-19 pandemic. The extent of COVID-19 involvement in the central nervous system is not well established, and the presence or the absence of SARS-CoV-2 particles in the cerebrospinal fluid (CSF) is a topic of debate. CASE DESCRIPTION: We present two patients with COVID-19 and concurrent neurological symptoms. Our first patient is a 31-year-old man who had flu-like symptoms due to COVID-19 and later developed an acute-onset severe headache and loss of consciousness and was diagnosed with a Hunt and Hess grade 3 subarachnoid haemorrhage from a ruptured aneurysm. Our second patient is a 62-year-old woman who had an ischaemic stroke with massive haemorrhagic conversion requiring a decompressive hemicraniectomy. Both patients' CSF was repeatedly negative on real-time PCR analysis despite concurrent neurological disease. CONCLUSION: Our report shows that patients' CSF may be devoid of viral particles even when they test positive for COVID-19 on a nasal swab. Whether SARS-CoV-2 is present in CSF may depend on the systemic disease severity and the degree of the virus' nervous tissue tropism and should be examined in future studies.


Subject(s)
Betacoronavirus/isolation & purification , Coronavirus Infections/cerebrospinal fluid , Coronavirus Infections/complications , Pneumonia, Viral/cerebrospinal fluid , Pneumonia, Viral/complications , Stroke/complications , Stroke/virology , Adult , COVID-19 , Female , Humans , Male , Middle Aged , Pandemics , SARS-CoV-2 , Stroke/cerebrospinal fluid
8.
Toxicol Appl Pharmacol ; 378: 114606, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31170415

ABSTRACT

Inorganic arsenic is an environmental carcinogen that poses a major global public health risk. A high percentage of drinking water from wells in the U.S. contains higher-than-normal levels of arsenic, suggesting an increased risk of arsenic-induced deleterious effects. In addition to primary preventive measures, therapeutic strategies need to effectively address and integrate multiple molecular mechanisms underlying arsenic-induced carcinogenesis. We previously showed that the loss of miR-199a-5p in arsenic-transformed cells is pivotal to promote arsenic-induced angiogenesis and tumor growth in lung epithelial cells. In this study, we further showed that subacute or chronic exposure to arsenic diminished miR-199a-5p levels largely due to DNA methylation, which was achieved by increased DNA methyltransferase-1 (DNMT1) activity, mediated by the formation of specific protein 1 (Sp1)/DNMT1 complex. In addition to the DNA hypermethylation, arsenic exposure also repressed miR-199a transcription through a transcriptional repressor Sp1. We further identified an association between miR-199a-5p repression and the arsenic-mediated energy metabolic shift, as reflected by mitochondria defects and a switch to glycolysis, in which a glycolytic enzyme pyruvate kinase 2 (PKM2) was a functional target of miR-199a-5p. Taken together, the repression of miR-199a-5p through both Sp1-dependent DNA methylation and Sp1 transcriptional repression promotes an arsenic-mediated metabolic shift from mitochondria respiration to aerobic glycolysis via PKM2.


Subject(s)
Arsenic/adverse effects , DNA Methylation/drug effects , MicroRNAs/genetics , Sp1 Transcription Factor/genetics , Activation, Metabolic/drug effects , Carcinogenesis/drug effects , Cell Line , Glycolysis/drug effects , Humans
9.
Am J Clin Oncol ; 42(5): 481-486, 2019 05.
Article in English | MEDLINE | ID: mdl-30973372

ABSTRACT

OBJECTIVES: Determine the prognostic significance of rapid early tumor progression before radiation and chemotherapy for glioblastoma patients. METHODS: A retrospective review of glioblastoma patients was performed. Rapid early progression (REP) was defined as new enhancing tumor or >25% increase in enhancement before radiotherapy. The pre/postoperative magnetic resonance imaging was compared with the preradiation magnetic resonance imaging to determine REP. A blinded review of imaging was performed. Kaplan-Meier curves were generated to compare progression-free and overall survival (OS). Univariate analysis was performed using the log-rank test for categorical variables and Cox proportional hazards for continuous variables. Multivariable logistic regression was performed to assess factors related to early progression and Cox proportional hazards model was used for multivariate analysis of OS. RESULTS: Eighty-seven patients met entry criteria. A total of 52% of patients developed REP. The OS in the REP group was 11.5 months (95% confidence interval [CI]: 7.4-17.6) and 20.1 months (95% CI: 17.8-26.1) without REP (P=0.013). On multivariate analysis including significant prognostic factors, presence of REP was found to increase the risk of death (hazard ratio: 2.104, 95% CI: 1.235-3.583, P=0.006). A total of 74% of patients recurred in the site of REP. CONCLUSIONS: REP was common and independently predicted for a worse OS. Integrating REP with MGMT promotor methylation improved prognostic assessment. The site of REP was a common site of tumor progression. Our findings are hypothesis generating and may indicate a particular subset of glioblastoma patients who are resistant to current standard of care therapy. Further study to determine other molecular features of this group are underway.


Subject(s)
Brain Neoplasms/mortality , Brain Neoplasms/pathology , Glioblastoma/mortality , Glioblastoma/pathology , Neurosurgical Procedures/methods , Adult , Aged , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Cohort Studies , Disease Progression , Disease-Free Survival , Female , Glioblastoma/diagnostic imaging , Glioblastoma/therapy , Humans , Kaplan-Meier Estimate , Logistic Models , Magnetic Resonance Imaging/methods , Male , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Staging , Prognosis , Proportional Hazards Models , Radiotherapy, Adjuvant , Retrospective Studies , Risk Assessment , Survival Analysis , Time Factors , United States
10.
Matrix Biol ; 77: 41-57, 2019 04.
Article in English | MEDLINE | ID: mdl-30098419

ABSTRACT

The αvß3 integrin has been shown to promote aggressive phenotypes in many types of cancers, including prostate cancer. We show that GFP-labeled αvß3 derived from cancer cells circulates in the blood and is detected in distant lesions in NOD scid gamma (NSG) mice. We, therefore, hypothesized that αvß3 travels through exosomes and tested its levels in pools of vesicles, which we designate extracellular vesicles highly enriched in exosomes (ExVs), and in exosomes isolated from the plasma of prostate cancer patients. Here, we show that the αvß3 integrin is found in patient blood exosomes purified by sucrose or iodixanol density gradients. In addition, we provide evidence that the αvß3 integrin is transferred through ExVs isolated from prostate cancer patient plasma to ß3-negative recipient cells. We also demonstrate the intracellular localization of ß3-GFP transferred via cancer cell-derived ExVs. We show that the ExVs present in plasma from prostate cancer patients contain higher levels of αvß3 and CD9 as compared to plasma ExVs from age-matched subjects who are not affected by cancer. Furthermore, using PSMA antibody-bead mediated immunocapture, we show that the αvß3 integrin is expressed in a subset of exosomes characterized by PSMA, CD9, CD63, and an epithelial-specific marker, Trop-2. Finally, we present evidence that the levels of αvß3, CD63, and CD9 remain unaltered in ExVs isolated from the blood of prostate cancer patients treated with enzalutamide. Our results suggest that detecting exosomal αvß3 integrin in prostate cancer patients could be a clinically useful and non-invasive biomarker to follow prostate cancer progression. Moreover, the ability of αvß3 integrin to be transferred from ExVs to recipient cells provides a strong rationale for further investigating the role of αvß3 integrin in the pathogenesis of prostate cancer and as a potential therapeutic target.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Exosomes/metabolism , Integrin alphaVbeta3/genetics , Prostatic Neoplasms/genetics , Adenocarcinoma/blood , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Benzamides , Biomarkers, Tumor/blood , Exosomes/chemistry , Gene Expression , Humans , Integrin alphaVbeta3/blood , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Nitriles , PC-3 Cells , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/blood , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Tetraspanin 29/blood , Tetraspanin 29/genetics , Tetraspanin 30/blood , Tetraspanin 30/genetics , Xenograft Model Antitumor Assays
11.
J Biol Chem ; 293(31): 12137-12148, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29921586

ABSTRACT

Regulation of autophagy by proteolytically cleaved fragments of heparan sulfate proteoglycans is a novel and current research focus in tumor biology. Endorepellin is the C-terminal angiostatic fragment of the heparan sulfate proteoglycan perlecan and induces autophagy in endothelial cells. To further investigate this property, we used NanoString, a digital PCR platform for measuring pre-defined transcripts in biological samples to analyze a custom subset of 95 autophagy-related genes in human umbilical vein endothelial cells treated with ultrapure human recombinant endorepellin. We discovered an endorepellin-evoked pro-autophagic and pro-mitophagic gene expression signatures, which included two coordinately up-regulated mitochondrial-associated genes encoding the E3 ubiquitin protein ligase Parkin and the tumor suppressor mitostatin. Induction of both proteins required the tyrosine kinase activity of vascular endothelial growth factor receptor 2 (VEGFR2). Furthermore, we discovered that endorepellin evoked mitochondrial depolarization in endothelial cells via a specific interaction between its two proximal LG1/2 domains and VEGFR2. We also found that following loss of membrane potential, mitostatin and parkin interact and that mitostatin associates with the established Parkin receptor mitofusin-2. In conclusion, we have identified a critical role for endorepellin in remodeling the autophagic transcriptome and influencing mitochondrial homeostasis.


Subject(s)
Autophagy , Heparan Sulfate Proteoglycans/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Mitophagy , Peptide Fragments/metabolism , Carrier Proteins , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Heparan Sulfate Proteoglycans/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Peptide Fragments/genetics , Protein Binding , Signal Transduction , Transcriptome , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
J Clin Oncol ; 36(4): 414-424, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29236593

ABSTRACT

Purpose Guidelines are limited for genetic testing for prostate cancer (PCA). The goal of this conference was to develop an expert consensus-driven working framework for comprehensive genetic evaluation of inherited PCA in the multigene testing era addressing genetic counseling, testing, and genetically informed management. Methods An expert consensus conference was convened including key stakeholders to address genetic counseling and testing, PCA screening, and management informed by evidence review. Results Consensus was strong that patients should engage in shared decision making for genetic testing. There was strong consensus to test HOXB13 for suspected hereditary PCA, BRCA1/2 for suspected hereditary breast and ovarian cancer, and DNA mismatch repair genes for suspected Lynch syndrome. There was strong consensus to factor BRCA2 mutations into PCA screening discussions. BRCA2 achieved moderate consensus for factoring into early-stage management discussion, with stronger consensus in high-risk/advanced and metastatic setting. Agreement was moderate to test all men with metastatic castration-resistant PCA, regardless of family history, with stronger agreement to test BRCA1/2 and moderate agreement to test ATM to inform prognosis and targeted therapy. Conclusion To our knowledge, this is the first comprehensive, multidisciplinary consensus statement to address a genetic evaluation framework for inherited PCA in the multigene testing era. Future research should focus on developing a working definition of familial PCA for clinical genetic testing, expanding understanding of genetic contribution to aggressive PCA, exploring clinical use of genetic testing for PCA management, genetic testing of African American males, and addressing the value framework of genetic evaluation and testing men at risk for PCA-a clinically heterogeneous disease.


Subject(s)
Biomarkers, Tumor/genetics , Genetic Testing/methods , Prostatic Neoplasms/genetics , Adult , Age Factors , Aged , Clinical Decision-Making , Genetic Predisposition to Disease , Genetic Testing/standards , Heredity , Humans , Male , Middle Aged , Pedigree , Phenotype , Predictive Value of Tests , Prognosis , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Risk Factors
14.
Cancer Biol Ther ; 19(4): 249-253, 2018 04 03.
Article in English | MEDLINE | ID: mdl-27466707

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) is a metabolic enzyme implicated in cancer cell metabolic reprogramming. This is underscored by the detection of functional, somatic IDH1 mutations frequently found in secondary glioblastoma. To our knowledge, there has never been a reported, validated case of an IDH1 mutation in a pancreatic ductal adenocarcinoma (PDA). Herein, we present a case of a patient with metastatic PDA that harbored a potentially actionable, albeit rare, IDH1 mutation. As part of the Know Your Tumor project (Pancreatic Cancer Action Network), a 48-year-old female was diagnosed with metastatic PDA and subsequently started on standard of care chemotherapy, during which her hepatic lesions progressed. Detailed molecular profiling was performed on a biopsy from a liver lesion that demonstrated an IDH1 mutation, R132H. This mutation was confirmed by an independent sequencing reaction from the tumor sample, and by immunohistochemistry using an antibody specific for the IDH1 R132H mutation. The patient subsequently received a mutant IDH1 inhibitor (AG-120, Agios Pharmaceuticals, Cambridge, MA), but with no response. IDH1 mutations are common in certain cancer types, but have not been reported in PDA. We report the first case of an IDH1 mutation in this tumor type, perhaps providing a rare opportunity for a targeted therapy as a treatment option for PDA.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/genetics , Isocitrate Dehydrogenase/genetics , Liver Neoplasms/genetics , Pancreatic Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Biopsy , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/secondary , DNA Mutational Analysis , Drug Resistance, Neoplasm , Female , Glycine/analogs & derivatives , Glycine/pharmacology , Glycine/therapeutic use , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Middle Aged , Mutation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pyridines/pharmacology , Pyridines/therapeutic use , Treatment Outcome
15.
Oncotarget ; 8(23): 37423-37434, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28445125

ABSTRACT

Intratumoral heterogeneity (ITH) is a prominent feature of kidney cancer. It is not known whether it has utility in finding associations between protein expression and clinical parameters. We used ITH that is detected by immunohistochemistry (IHC) to aid the association analysis between the loss of SWI/SNF components and clinical parameters.160 ccRCC tumors (40 per tumor stage) were used to generate tissue microarray (TMA). Four foci from different regions of each tumor were selected. IHC was performed against PBRM1, ARID1A, SETD2, SMARCA4, and SMARCA2. Statistical analyses were performed to correlate biomarker losses with patho-clinical parameters. Categorical variables were compared between groups using Fisher's exact tests. Univariate and multivariable analyses were used to correlate biomarker changes and patient survivals. Multivariable analyses were performed by constructing decision trees using the classification and regression trees (CART) methodology. IHC detected widespread ITH in ccRCC tumors. The statistical analysis of the "Truncal loss" (root loss) found additional correlations between biomarker losses and tumor stages than the traditional "Loss in tumor (total)". Losses of SMARCA4 or SMARCA2 significantly improved prognosis for overall survival (OS). Losses of PBRM1, ARID1A or SETD2 had the opposite effect. Thus "Truncal Loss" analysis revealed hidden links between protein losses and patient survival in ccRCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Proteins/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/pathology , DNA Helicases/metabolism , DNA-Binding Proteins , Female , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Kidney Neoplasms/pathology , Male , Middle Aged , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Young Adult
16.
Matrix Biol ; 64: 27-39, 2017 12.
Article in English | MEDLINE | ID: mdl-28433812

ABSTRACT

Despite extensive clinical and experimental studies over the past decades, the pathogenesis and progression to the castration-resistant stage of prostate cancer remains largely unknown. Progranulin, a secreted growth factor, strongly binds the heparin-sulfate proteoglycan perlecan, and counteracts its biological activity. We established that progranulin acts as an autocrine growth factor and promotes prostate cancer cell motility, invasion, and anchorage-independent growth. Progranulin was overexpressed in prostate cancer tissues vis-à-vis non-neoplastic tissues supporting the hypothesis that progranulin may play a key role in prostate cancer progression. However, progranulin's mode of action is not well understood and proteins regulating progranulin signaling have not been identified. Sortilin, a single-pass type I transmembrane protein of the Vps10 family, binds progranulin in neurons and targets progranulin for lysosomal degradation. Significantly, in DU145 and PC3 cells, we detected very low levels of sortilin associated with high levels of progranulin production and enhanced motility. Restoring sortilin expression decreased progranulin levels, inhibited motility and anchorage-independent growth and destabilized Akt. These results demonstrated a critical role for sortilin in regulating progranulin and suggest that sortilin loss may contribute to prostate cancer progression. Here, we provide the novel observation that progranulin downregulated sortilin protein levels independent of transcription. Progranulin induced sortilin ubiquitination, internalization via clathrin-dependent endocytosis and sorting into early endosomes for lysosomal degradation. Collectively, these results constitute a regulatory feed-back mechanism whereby sortilin downregulation ensures sustained progranulin-mediated oncogenesis.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Autocrine Communication , Cell Line, Tumor , Cell Movement , Down-Regulation , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Humans , Male , Progranulins , Prostatic Neoplasms, Castration-Resistant/genetics , Protein Transport , Proteolysis , Transcription, Genetic , Ubiquitination
17.
BMC Cancer ; 16(1): 766, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27716119

ABSTRACT

BACKGROUND: We sought to assess physician interest in molecular prognosic testing for patients with early stage colon cancer, and identify factors associated with the likelihood of test adoption. METHODS: We identified physicians who care for patients with early-stage (pN0) colon cancer patients, mailed them a survey, and analyzed survey responses to assess clinician receptivity to the use of a new molecular test (GUCY2C) that identifies patients at risk for recurrence, and clinician readiness to act on abnormal test results. RESULTS: Of 104 eligible potential respondents, 41 completed and returned the survey. Among responding physicians, 56 % were receptive to using the new prognostic test. Multivariable analyses showed that physicians in academic medical centers were significantly more receptive to molecular test use than those in non-academic settings. Forty-one percent of respondents were ready to act on abnormal molecular test results. Physicians who viewed current staging methods as inaccurate and were confident in their capacity to incorporate molecular testing in practice were more likely to say they would act on abnormal test results. CONCLUSIONS: Physician receptivity to molecular diagnostic testing for early-stage colon cancer patients is likely to be influenced by practice setting and perceptions related to delivering quality care to patients. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT01972737.


Subject(s)
Colonic Neoplasms/diagnosis , Molecular Diagnostic Techniques , Practice Patterns, Physicians' , Female , Health Care Surveys , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/prevention & control , Neoplasm Staging , Prognosis , Risk Assessment , Risk Factors
18.
PLoS One ; 11(10): e0164554, 2016.
Article in English | MEDLINE | ID: mdl-27764136

ABSTRACT

Recent studies have shown that intratumoral heterogeneity (ITH) is prevalent in clear cell renal cell carcinoma (ccRCC), based on DNA sequencing and chromosome aberration analysis of multiple regions from the same tumor. VHL mutations were found to be universal throughout individual tumors when it occurred (ubiquitous), while the mutations in other tumor suppressor genes tended to be detected only in parts of the tumors (subclonal). ITH has been studied mostly by DNA sequencing in limited numbers of samples, either by whole genome sequencing or by targeted sequencing. It is not known whether immunohistochemistry (IHC) can be used as a tool to study ITH. To address this question, we examined the protein expression of PBRM1, and PBRM1-related proteins such as ARID1A, SETD2, BRG1, and BRM. Altogether, 160 ccRCC (40 per stage) were used to generate a tissue microarray (TMA), with four foci from each tumor included. Loss of expression was defined as 0-5% of tumor cells with positive nuclear staining in an individual focus. We found that 49/160 (31%), 81/160 (51%), 23/160 (14%), 24/160 (15%), and 61/160 (38%) of ccRCC showed loss of expression of PBRM1, ARID1A, SETD2, BRG1, and BRM, respectively, and that IHC could successfully detect a high prevalence of ITH. Phylogenetic trees were constructed that reflected the ITH. Striking co-losses among proteins were also observed. For instance, ARID1A loss almost always accompanied PBRM1 loss, whereas BRM loss accompanied loss of BRG1, PBRM1 or ARID1A. SETD2 loss frequently occurred with loss of one or more of the other four proteins. Finally, in order to learn the impact of combined losses, we compared the tumor growth after cells acquired losses of ARID1A, PBRM1, or both in a xenograft model. The results suggest that ARID1A loss has a greater tumor-promoting effect than PBRM1 loss, indicating that xenograft analysis is a useful tool to investigate how these losses impact on tumor behavior, either alone or in combination.


Subject(s)
Carcinoma, Renal Cell/metabolism , Chromatin/metabolism , Gene Deletion , Kidney Neoplasms/metabolism , Neoplasm Proteins/metabolism , Tissue Array Analysis/methods , Animals , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , DNA Helicases/metabolism , DNA-Binding Proteins , Down-Regulation , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunohistochemistry , Kidney Neoplasms/pathology , Mice , Nuclear Proteins/metabolism , Phylogeny , Transcription Factors/metabolism
19.
Oncotarget ; 7(26): 39980-39995, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27220888

ABSTRACT

We have recently demonstrated a critical role for progranulin in bladder cancer. Progranulin contributes, as an autocrine growth factor, to the transformed phenotype by modulating Akt-and MAPK-driven motility, invasion and anchorage-independent growth. Progranulin also induces F-actin remodeling by interacting with the F-actin binding protein drebrin. In addition, progranulin is overexpressed in invasive bladder cancer compared to normal tissue controls, suggesting that progranulin might play a key role in driving the transition to the invasive phenotype of urothelial cancer. However, it is not established whether targeting progranulin could have therapeutic effects on bladder cancer. In this study, we stably depleted urothelial cancer cells of endogenous progranulin by shRNA approaches and determined that progranulin depletion severely inhibited the ability of tumorigenic urothelial cancer cells to migrate, invade and grow in anchorage-independency. We further demonstrate that progranulin expression is critical for tumor growth in vivo, in both xenograft and orthotopic tumor models. Notably, progranulin levels correlated with response to cisplatin treatment and were upregulated in bladder tumors. Our data indicate that progranulin may constitute a novel target for therapeutic intervention in bladder tumors. In addition, progranulin may serve as a novel biomarker for bladder cancer.


Subject(s)
Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Actins/metabolism , Animals , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Female , Humans , MAP Kinase Signaling System , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Invasiveness , Neoplasm Transplantation , Phenotype , Progranulins , RNA, Small Interfering/metabolism , Urothelium/pathology
20.
Oncotarget ; 6(13): 10825-39, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25839164

ABSTRACT

We recently established a critical role for the growth factor progranulin in bladder cancer insofar as progranulin promotes urothelial cancer cell motility and contributes, as an autocrine growth factor, to the transformed phenotype by modulating invasion and anchorage-independent growth. In addition, progranulin expression is upregulated in invasive bladder cancer tissues compared to normal controls. However, the molecular mechanisms of progranulin action in bladder cancer have not been fully elucidated. In this study, we searched for novel progranulin-interacting proteins using pull-down assays with recombinant progranulin and proteomics. We discovered that drebrin, an F-actin binding protein, bound progranulin in urothelial cancer cells. We characterized drebrin function in urothelial cancer cell lines and showed that drebrin is critical for progranulin-dependent activation of the Akt and MAPK pathways and modulates motility, invasion and anchorage-independent growth. In addition, drebrin regulates tumor formation in vivo and its expression is upregulated in bladder cancer tissues compared to normal tissue controls. Our data are translationally relevant as indicate that drebrin exerts an essential functional role in the regulation of progranulin action and may constitute a novel target for therapeutic intervention in bladder tumors. In addition, drebrin may serve as novel biomarker for bladder cancer.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Urinary Bladder Neoplasms/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Neuropeptides/genetics , Progranulins , Protein Binding , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction , Transfection , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urothelium/metabolism , Urothelium/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...