Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Thorac Cancer ; 13(21): 3032-3041, 2022 11.
Article in English | MEDLINE | ID: mdl-36101520

ABSTRACT

BACKGROUND: ROS1 tyrosine kinase inhibitors (TKIs) have demonstrated significant clinical benefit for ROS1+ NSCLC patients. However, TKI resistance inevitably develops through ROS1 kinase domain (KD) modification or another kinase driving bypass signaling. While multiple TKIs have been designed to target ROS1 KD mutations, less is known about bypass signaling in TKI-resistant ROS1+ lung cancers. METHODS: Utilizing a primary, patient-derived TPM3-ROS1 cell line (CUTO28), we derived an entrectinib-resistant line (CUTO28-ER). We evaluated proliferation and signaling responses to TKIs, and utilized RNA sequencing, whole exome sequencing, and fluorescence in situ hybridization to detect transcriptional, mutational, and copy number alterations, respectively. We substantiated in vitro findings using a CD74-ROS1 NSCLC patient's tumor samples. Last, we analyzed circulating tumor DNA (ctDNA) from ROS1+ NSCLC patients in the STARTRK-2 entrectinib trial to determine the prevalence of MET amplification. RESULTS: CUTO28-ER cells did not exhibit ROS1 KD mutations. MET TKIs inhibited proliferation and downstream signaling and MET transcription was elevated in CUTO28-ER cells. CUTO28-ER cells displayed extrachromosomal (ecDNA) MET amplification without MET activating mutations, exon 14 skipping, or fusions. The CD74-ROS1 patient samples illustrated MET amplification while receiving ROS1 TKI. Finally, two of 105 (1.9%) entrectinib-resistant ROS1+ NSCLC STARTRK-2 patients with ctDNA analysis at enrollment and disease progression displayed MET amplification. CONCLUSIONS: Treatment with ROS1-selective inhibitors may lead to MET-mediated resistance. The discovery of ecDNA MET amplification is noteworthy, as ecDNA is associated with more aggressive cancers. Following progression on ROS1-selective inhibitors, MET gene testing and treatments targeting MET should be explored to overcome MET-driven resistance.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm/genetics , Gene Amplification , In Situ Hybridization, Fluorescence , Lung Neoplasms/pathology , Mutation , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Clinical Trials as Topic
2.
Nat Commun ; 11(1): 4374, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32873787

ABSTRACT

Oncogene amplification, a major driver of cancer pathogenicity, is often mediated through focal amplification of genomic segments. Recent results implicate extrachromosomal DNA (ecDNA) as the primary driver of focal copy number amplification (fCNA) - enabling gene amplification, rapid tumor evolution, and the rewiring of regulatory circuitry. Resolving an fCNA's structure is a first step in deciphering the mechanisms of its genesis and the fCNA's subsequent biological consequences. We introduce a computational method, AmpliconReconstructor (AR), for integrating optical mapping (OM) of long DNA fragments (>150 kb) with next-generation sequencing (NGS) to resolve fCNAs at single-nucleotide resolution. AR uses an NGS-derived breakpoint graph alongside OM scaffolds to produce high-fidelity reconstructions. After validating its performance through multiple simulation strategies, AR reconstructed fCNAs in seven cancer cell lines to reveal the complex architecture of ecDNA, a breakage-fusion-bridge and other complex rearrangements. By reconstructing the rearrangement signatures associated with an fCNA's generative mechanism, AR enables a more thorough understanding of the origins of fCNAs.


Subject(s)
Gene Amplification , Genomics/methods , Neoplasms/genetics , Oncogenes/genetics , Cell Line, Tumor , Chromosome Mapping/methods , Cytogenetic Analysis , Genome, Human/genetics , High-Throughput Nucleotide Sequencing/methods , Humans
4.
Nat Commun ; 11(1): 550, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992716

ABSTRACT

Many cellular models aimed at elucidating cancer biology do not recapitulate pathobiology including tumor heterogeneity, an inherent feature of cancer that underlies treatment resistance. Here we introduce a cancer modeling paradigm using genetically engineered human pluripotent stem cells (hiPSCs) that captures authentic cancer pathobiology. Orthotopic engraftment of the neural progenitor cells derived from hiPSCs that have been genome-edited to contain tumor-associated genetic driver mutations revealed by The Cancer Genome Atlas project for glioblastoma (GBM) results in formation of high-grade gliomas. Similar to patient-derived GBM, these models harbor inter-tumor heterogeneity resembling different GBM molecular subtypes, intra-tumor heterogeneity, and extrachromosomal DNA amplification. Re-engraftment of these primary tumor neurospheres generates secondary tumors with features characteristic of patient samples and present mutation-dependent patterns of tumor evolution. These cancer avatar models provide a platform for comprehensive longitudinal assessment of human tumor development as governed by molecular subtype mutations and lineage-restricted differentiation.


Subject(s)
Genetic Engineering , Glioblastoma/genetics , Glioblastoma/pathology , Pluripotent Stem Cells/pathology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Genome , Glioblastoma/metabolism , Glioma/genetics , Glioma/pathology , Humans , Mice , Mice, SCID , Mutation , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Neurofibromin 1/genetics , PTEN Phosphohydrolase/genetics , Transplantation, Heterologous , Tumor Suppressor Protein p53/genetics
5.
Mol Cancer Ther ; 19(2): 602-613, 2020 02.
Article in English | MEDLINE | ID: mdl-31575654

ABSTRACT

Patients harboring germline breast cancer susceptibility genes 1 and 2 (BRCA1/2) mutations are predisposed to developing breast, pancreatic, and ovarian cancers. BRCA2 plays a critical role in homologous recombination (HR) DNA repair and deleterious mutations in BRCA2 confer sensitivity to PARP inhibition. Recently, the PARP inhibitors olaparib and rucaparib were FDA approved for the treatment of metastatic breast cancer and patients with recurrent ovarian cancer with mutations in BRCA1/2. Despite their initial antitumor activity, the development of resistance limits the clinical utility of PARP inhibitor therapy. Multiple resistance mechanisms have been described, including reversion mutations that restore the reading frame of the BRCA2 gene. In this study, we generated olaparib- and rucaparib-resistant BRCA2-mutant Capan1 cell lines. We did not detect secondary reversion mutations in the olaparib- or rucaparib-resistant clones. Several of the resistant clones had gene duplication and amplification of the mutant BRCA2 allele, with a corresponding increase in expression of a truncated BRCA2 protein. In addition, HR-mediated DNA repair was rescued, as evidenced by the restoration of RAD51 foci formation. Using mass spectrometry, we identified Disruptor Of Telomeric silencing 1-Like (DOT1L), as an interacting partner of truncated BRCA2. RNAi-mediated knockdown of BRCA2 or DOT1L was sufficient to resensitize cells to olaparib. The results demonstrate that independent of a BRCA2 reversion, mutation amplification of a mutant-carrying BRCA2 contributes to PARP inhibitor resistance.


Subject(s)
BRCA2 Protein/metabolism , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Rad51 Recombinase/metabolism , Cell Line, Tumor , Female , Humans , Mutation
6.
Nature ; 575(7784): 699-703, 2019 11.
Article in English | MEDLINE | ID: mdl-31748743

ABSTRACT

Oncogenes are commonly amplified on particles of extrachromosomal DNA (ecDNA) in cancer1,2, but our understanding of the structure of ecDNA and its effect on gene regulation is limited. Here, by integrating ultrastructural imaging, long-range optical mapping and computational analysis of whole-genome sequencing, we demonstrate the structure of circular ecDNA. Pan-cancer analyses reveal that oncogenes encoded on ecDNA are among the most highly expressed genes in the transcriptome of the tumours, linking increased copy number with high transcription levels. Quantitative assessment of the chromatin state reveals that although ecDNA is packaged into chromatin with intact domain structure, it lacks higher-order compaction that is typical of chromosomes and displays significantly enhanced chromatin accessibility. Furthermore, ecDNA is shown to have a significantly greater number of ultra-long-range interactions with active chromatin, which provides insight into how the structure of circular ecDNA affects oncogene function, and connects ecDNA biology with modern cancer genomics and epigenetics.


Subject(s)
Chromatin/genetics , DNA, Circular/metabolism , Gene Expression Regulation, Neoplastic/genetics , Neoplasms/genetics , Oncogenes/genetics , Cell Line, Tumor , Chromatin/chemistry , DNA, Circular/genetics , Humans , Microscopy, Electron, Scanning , Neoplasms/physiopathology
7.
Cell Metab ; 30(3): 525-538.e8, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31303424

ABSTRACT

Advances in DNA sequencing technologies have reshaped our understanding of the molecular basis of cancer, providing a precise genomic view of tumors. Complementary biochemical and biophysical perspectives of cancer point toward profound shifts in nutrient uptake and utilization that propel tumor growth and major changes in the structure of the plasma membrane of tumor cells. The molecular mechanisms that bridge these fundamental aspects of tumor biology remain poorly understood. Here, we show that the lysophosphatidylcholine acyltransferase LPCAT1 functionally links specific genetic alterations in cancer with aberrant metabolism and plasma membrane remodeling to drive tumor growth. Growth factor receptor-driven cancers are found to depend on LPCAT1 to shape plasma membrane composition through enhanced saturated phosphatidylcholine content that is, in turn, required for the transduction of oncogenic signals. These results point to a genotype-informed strategy that prioritizes lipid remodeling pathways as therapeutic targets for diverse cancers.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Gene Amplification , Neoplasms/genetics , Neoplasms/metabolism , Oncogenes/genetics , Phospholipids/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , A549 Cells , Animals , Cell Survival/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Genotype , Heterografts , Humans , Mice , Mice, Nude , PC-3 Cells , Signal Transduction/genetics , Transfection
8.
Nat Commun ; 10(1): 392, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30674876

ABSTRACT

Focal oncogene amplification and rearrangements drive tumor growth and evolution in multiple cancer types. We present AmpliconArchitect (AA), a tool to reconstruct the fine structure of focally amplified regions using whole genome sequencing (WGS) and validate it extensively on multiple simulated and real datasets, across a wide range of coverage and copy numbers. Analysis of AA-reconstructed amplicons in a pan-cancer dataset reveals many novel properties of copy number amplifications in cancer. These findings support a model in which focal amplifications arise due to the formation and replication of extrachromosomal DNA. Applying AA to 68 viral-mediated cancer samples, we identify a large fraction of amplicons with specific structural signatures suggestive of hybrid, human-viral extrachromosomal DNA. AA reconstruction, integrated with metaphase fluorescence in situ hybridization (FISH) and PacBio sequencing on the cell-line UPCI:SCC090 confirm the extrachromosomal origin and fine structure of a Forkhead box E1 (FOXE1)-containing hybrid amplicon.


Subject(s)
Gene Amplification , Neoplasms/genetics , Algorithms , Cell Line , Cell Line, Tumor , Chromosome Duplication , Chromosomes, Human/genetics , Computers, Molecular , Forkhead Transcription Factors/genetics , Genes, Viral , Humans , In Situ Hybridization, Fluorescence
9.
Nature ; 543(7643): 122-125, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28178237

ABSTRACT

Human cells have twenty-three pairs of chromosomes. In cancer, however, genes can be amplified in chromosomes or in circular extrachromosomal DNA (ecDNA), although the frequency and functional importance of ecDNA are not understood. We performed whole-genome sequencing, structural modelling and cytogenetic analyses of 17 different cancer types, including analysis of the structure and function of chromosomes during metaphase of 2,572 dividing cells, and developed a software package called ECdetect to conduct unbiased, integrated ecDNA detection and analysis. Here we show that ecDNA was found in nearly half of human cancers; its frequency varied by tumour type, but it was almost never found in normal cells. Driver oncogenes were amplified most commonly in ecDNA, thereby increasing transcript level. Mathematical modelling predicted that ecDNA amplification would increase oncogene copy number and intratumoural heterogeneity more effectively than chromosomal amplification. We validated these predictions by quantitative analyses of cancer samples. The results presented here suggest that ecDNA contributes to accelerated evolution in cancer.


Subject(s)
DNA Copy Number Variations/genetics , Evolution, Molecular , Gene Amplification/genetics , Genetic Heterogeneity , Models, Genetic , Neoplasms/genetics , Oncogenes/genetics , Chromosomes, Human/genetics , Cytogenetic Analysis , DNA Mutational Analysis , Genome, Human/genetics , Humans , Metaphase/genetics , Neoplasms/classification , RNA, Messenger/analysis , RNA, Neoplasm/genetics , Reproducibility of Results , Software
10.
Cancer Cell ; 30(5): 683-693, 2016 Nov 14.
Article in English | MEDLINE | ID: mdl-27746144

ABSTRACT

Small-molecule inhibitors targeting growth factor receptors have failed to show efficacy for brain cancers, potentially due to their inability to achieve sufficient drug levels in the CNS. Targeting non-oncogene tumor co-dependencies provides an alternative approach, particularly if drugs with high brain penetration can be identified. Here we demonstrate that the highly lethal brain cancer glioblastoma (GBM) is remarkably dependent on cholesterol for survival, rendering these tumors sensitive to Liver X receptor (LXR) agonist-dependent cell death. We show that LXR-623, a clinically viable, highly brain-penetrant LXRα-partial/LXRß-full agonist selectively kills GBM cells in an LXRß- and cholesterol-dependent fashion, causing tumor regression and prolonged survival in mouse models. Thus, a metabolic co-dependency provides a pharmacological means to kill growth factor-activated cancers in the CNS.


Subject(s)
Brain Neoplasms/drug therapy , Cholesterol/metabolism , Glioblastoma/drug therapy , Indazoles/administration & dosage , Liver X Receptors/metabolism , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Glioblastoma/metabolism , Humans , Indazoles/pharmacology , Mice , Treatment Outcome
11.
Mol Cell ; 60(2): 307-18, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26455392

ABSTRACT

Epidermal growth factor receptor (EGFR) gene amplification and mutations are the most common oncogenic events in glioblastoma (GBM), but the mechanisms by which they promote aggressive tumor growth are not well understood. Here, through integrated epigenome and transcriptome analyses of cell lines, genotyped clinical samples, and TCGA data, we show that EGFR mutations remodel the activated enhancer landscape of GBM, promoting tumorigenesis through a SOX9 and FOXG1-dependent transcriptional regulatory network in vitro and in vivo. The most common EGFR mutation, EGFRvIII, sensitizes GBM cells to the BET-bromodomain inhibitor JQ1 in a SOX9, FOXG1-dependent manner. These results identify the role of transcriptional/epigenetic remodeling in EGFR-dependent pathogenesis and suggest a mechanistic basis for epigenetic therapy.


Subject(s)
Brain Neoplasms/genetics , Epigenesis, Genetic , ErbB Receptors/genetics , Forkhead Transcription Factors/genetics , Glioblastoma/genetics , Nerve Tissue Proteins/genetics , SOX9 Transcription Factor/genetics , Adult , Animals , Azepines/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Child , ErbB Receptors/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Nerve Tissue Proteins/metabolism , SOX9 Transcription Factor/metabolism , Signal Transduction , Transcriptome , Triazoles/pharmacology
13.
Proc Natl Acad Sci U S A ; 112(11): 3421-6, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25737557

ABSTRACT

Akt is a robust oncogene that plays key roles in the development and progression of many cancers, including glioma. We evaluated the differential propensities of the Akt isoforms toward progression in the well-characterized RCAS/Ntv-a mouse model of PDGFB-driven low grade glioma. A constitutively active myristoylated form of Akt1 did not induce high-grade glioma (HGG). In stark contrast, Akt2 and Akt3 showed strong progression potential with 78% and 97% of tumors diagnosed as HGG, respectively. We further revealed that significant variations in polarity and hydropathy values among the Akt isoforms in both the pleckstrin homology domain (P domain) and regulatory domain (R domain) were critical in mediating glioma progression. Gene expression profiles from representative Akt-derived tumors indicated dominant and distinct roles for Akt3, consisting primarily of DNA repair pathways. TCGA data from human GBM closely reflected the DNA repair function, as Akt3 was significantly correlated with a 76-gene signature DNA repair panel. Consistently, compared with Akt1 and Akt2 overexpression models, Akt3-expressing human GBM cells had enhanced activation of DNA repair proteins, leading to increased DNA repair and subsequent resistance to radiation and temozolomide. Given the wide range of Akt3-amplified cancers, Akt3 may represent a key resistance factor.


Subject(s)
Brain Neoplasms/genetics , DNA Repair/genetics , Disease Progression , Gene Amplification , Genome, Human , Glioma/genetics , Proto-Oncogene Proteins c-akt/genetics , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Line, Tumor , DNA Damage/genetics , DNA Repair/drug effects , DNA Repair/radiation effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/radiation effects , Gene Amplification/drug effects , Gene Amplification/radiation effects , Gene Expression Regulation, Neoplastic , Glioma/enzymology , Glioma/pathology , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Radiation Tolerance/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/radiation effects , Temozolomide , Transcription, Genetic
14.
Neuro Oncol ; 16(10): 1313-23, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24811392

ABSTRACT

Effective cancer treatment has been limited by the emergence of resistant cancer cells. The results of many studies indicate that AKT activation plays an important role in the acquisition of resistance to anticancer therapy. AKT is a critical effector serine/threonine kinase in the receptor tyrosine kinase/phosphatase and tensin homolog/phospho-inositide 3-kinase pathway and controls a myriad of cellular functions. Activation of AKT not only supports tumor growth and progression but also contributes to tumor-cell evasion of the cytotoxic effects of cancer therapy through many avenues including the promotion of anti-apoptosis, proliferation, and migration and regulation of the cell cycle. Accumulating evidence has implicated AKT as a direct participant in the DNA damage response and repair induced by commonly used genotoxic agents. In this review, we discuss the molecular mechanisms by which genotoxic agents activate AKT and therefore contribute to resistance to cancer therapeutics, with particular emphasis on DNA repair.


Subject(s)
DNA Repair , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Animals , DNA Damage , Humans , Mice , Signal Transduction
15.
Pediatrics ; 133(3): e609-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24488749

ABSTRACT

BACKGROUND AND OBJECTIVES: Congenital cytomegalovirus (CMV) is the leading cause of nongenetic deafness in children in the United States and can cause neurodevelopmental impairment in term infants. Limited data exist regarding congenital CMV infections in preterm infants. We aimed to determine the incidence and association with outcomes of congenital CMV in very low birth weight (VLBW) preterm infants. METHODS: VLBW infants born in 1993 to 2008 and admitted to the University of Alabama in Birmingham Regional Neonatal ICU were screened on admission for congenital CMV. CMV status and clinical outcomes were identified by using internal patient databases and hospital-based medical records. The primary outcome was death. Secondary outcomes included evidence of neurologic injury in the form of abnormal cranial ultrasound findings, sensorineural hearing loss, or abnormal motor development. Multivariate analysis was performed. RESULTS: Eighteen of 4594 VLBW infants had congenital CMV (0.39%; 95% confidence interval, 0.25%-0.62%). An additional 16 infants (0.35%; 95% confidence interval, 0.21%-0.57%) were identified who acquired CMV postnatally. Congenital CMV was not associated with death. Compared with controls, congenitally infected VLBW infants were more likely to have hearing loss at initial screening (67% vs. 9%, P < .0001) and confirmed at follow-up (83% vs. 2.1%, P < .0001). Congenital CMV was also associated with abnormal neuroimaging (72% vs. 25%, P < .0001) and adverse developmental motor outcomes (43% vs. 9%, P = .02). Acquired CMV was not associated with any adverse outcomes. CONCLUSIONS: Congenital CMV in VLBW infants is associated with high rates of neurologic injury and hearing loss but not death.


Subject(s)
Cytomegalovirus Infections/epidemiology , Hearing Loss, Sensorineural/epidemiology , Infant, Very Low Birth Weight , Neonatal Screening , Nervous System Diseases/epidemiology , Pregnancy Complications, Infectious/epidemiology , Adult , Cytomegalovirus Infections/diagnosis , Female , Follow-Up Studies , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/virology , Humans , Incidence , Infant, Newborn , Infant, Very Low Birth Weight/growth & development , Neonatal Screening/methods , Nervous System Diseases/diagnosis , Nervous System Diseases/virology , Pregnancy , Pregnancy Complications, Infectious/diagnosis , Retrospective Studies , Young Adult
16.
J Cardiovasc Magn Reson ; 15: 108, 2013 Dec 21.
Article in English | MEDLINE | ID: mdl-24359617

ABSTRACT

BACKGROUND: Regadenoson is a vasodilator stress agent that selectively activates the A2A receptor. Compared to adenosine, regadenoson is easier to administer and results in fewer side effects. Although extensively studied in patients undergoing nuclear perfusion imaging (MPI), its use for perfusion cardiovascular magnetic resonance (CMR) is not well described. The aim of this study was to determine the prognostic value of a normal regadenoson perfusion CMR in patients with known or suspected coronary artery disease. METHODS: Patients with known or suspected coronary artery disease were prospectively enrolled to receive perfusion CMR (Philips 1.5 T) with regadenoson. Three short-axis slices of the left ventricle (LV) were obtained during first pass of contrast using a hybrid GRE-EPI pulse sequence (0.075 mmol/kg Gadolinium-DTPA-BMA at 4 ml/sec). Imaging was performed 1 minute after injection of regadenoson (0.4 mg) and repeated 15 minutes after reversal of hyperemia with aminophylline (125 mg). Perfusion defects were documented if they persisted for ≥ 2 frames after peak enhancement of the LV cavity. CMR was considered abnormal if there was a resting wall motion abnormality, decreased LVEF (<40%), presence of LGE, or the presence of a perfusion defect during hyperemia. All patients were followed for a minimum of 1 year for major adverse cardiovascular event (MACE) defined as coronary revascularization, non-fatal myocardial infarction, and cardiovascular death. RESULTS: 149 patients were included in the final analysis. Perfusion defects were noted in 43/149 (29%) patients; 59/149 (40%) had any abnormality on CMR. During the mean follow-up period of 24 ± 9 months, 17/149 (11.4%) patients experienced MACE. The separation in the survival distributions for those with perfusion defects and those without perfusion defects was highly significant (log-rank p = 0.0001). When the absence of perfusion defects was added to the absence of other resting CMR abnormalities, the negative predictive value improved from 96% to 99%. CONCLUSION: Regadenoson perfusion CMR provides high confidence for excellent prognosis in patients with normal perfusion.


Subject(s)
Coronary Artery Disease/diagnosis , Magnetic Resonance Imaging, Cine , Myocardial Perfusion Imaging/methods , Purines , Pyrazoles , Vasodilator Agents , Adult , Aged , Contrast Media , Coronary Artery Disease/complications , Coronary Artery Disease/mortality , Coronary Artery Disease/physiopathology , Coronary Artery Disease/therapy , Female , Gadolinium DTPA , Hemodynamics , Humans , Infusions, Intravenous , Kaplan-Meier Estimate , Male , Middle Aged , Myocardial Infarction , Myocardial Revascularization , Perfusion , Predictive Value of Tests , Prognosis , Purines/administration & dosage , Purines/adverse effects , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Risk Factors , Stroke Volume , Time Factors , Vasodilator Agents/administration & dosage , Vasodilator Agents/adverse effects , Ventricular Function, Left
17.
Pediatrics ; 125(6): e1505-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20478935

ABSTRACT

Paraquat is an herbicide that is highly toxic to humans. Pediatric ingestion has become uncommon in the United States because of preventative efforts. We report here an unintentional, fatal paraquat ingestion by an 8-year-old child. Storage in an inappropriate container, confusion between herbicide trade names, nonspecific symptoms, and a delay in follow-up produced challenges in the diagnosis. In the absence of a clear history of ingestion, paraquat poisoning should be suspected in children who develop skin and mucous membrane burns, gastrointestinal symptoms, acute kidney injury, and respiratory failure.


Subject(s)
Acute Lung Injury/chemically induced , Herbicides/poisoning , Paraquat/poisoning , Respiratory Insufficiency/chemically induced , Acute Lung Injury/pathology , Acute Lung Injury/physiopathology , Chest Wall Oscillation , Child , Disease Progression , Fatal Outcome , Humans , Hypoxia/chemically induced , Lung/drug effects , Lung/pathology , Male , Poisoning/diagnosis , Poisoning/physiopathology , Poisoning/therapy , Renal Insufficiency/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...