Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Oncotarget ; 6(31): 31844-56, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26378811

ABSTRACT

Pilocytic astrocytoma (PA) is the most common brain tumor in children but is rare in adults, and hence poorly studied in this age group. We investigated 222 PA and report increased aneuploidy in older patients. Aneuploid genomes were identified in 45% of adult compared with 17% of pediatric PA. Gains were non-random, favoring chromosomes 5, 7, 6 and 11 in order of frequency, and preferentially affecting non-cerebellar PA and tumors with BRAF V600E mutations and not with KIAA1549-BRAF fusions or FGFR1 mutations. Aneuploid PA differentially expressed genes involved in CNS development, the unfolded protein response, and regulators of genomic stability and the cell cycle (MDM2, PLK2),whose correlated programs were overexpressed specifically in aneuploid PA compared to other glial tumors. Thus, convergence of pathways affecting the cell cycle and genomic stability may favor aneuploidy in PA, possibly representing an additional molecular driver in older patients with this brain tumor.


Subject(s)
Aneuploidy , Astrocytoma/classification , Biomarkers, Tumor/genetics , Brain Neoplasms/classification , Adult , Age Factors , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Child , Cohort Studies , Female , Gene Expression Profiling , Humans , Male , Mutation/genetics , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
2.
Acta Neuropathol Commun ; 1: 17, 2013 May 20.
Article in English | MEDLINE | ID: mdl-24252689

ABSTRACT

BACKGROUND: Pilocytic astrocytomas occur predominantly in childhood. In contrast to the posterior fossa location, hypothalamo-chiasmatic pilocytic astrocytomas display a worse prognosis often leading to multiple surgical procedures and/or several lines of chemotherapy and radiotherapy to achieve long-term control. Hypothalamo-chiasmatic pilocytic astrocytomas and cerebellar pilocytic astrocytomas have a distinctive gene signature and several differential expressed genes (ICAM1, CRK, CD36, and IQGAP1) are targets for available drugs: fluvastatin and/or celecoxib. RESULTS: Quantification by RT-Q-PCR of the expression of these genes was performed in a series of 51 pilocytic astrocytomas and 10 glioblastomas: they were all significantly overexpressed in hypothalamo-chiasmatic pilocytic astrocytomas relative to cerebellar pilocytic astrocytomas, and CRK and ICAM1 were significantly overexpressed in pilocytic astrocytomas versus glioblastomas.We used two commercially available glioblastoma cell lines and three pilocytic astrocytoma explant cultures to investigate the effect of celecoxib/fluvastatin alone or in combination. Glioblastoma cell lines were sensitive to both drugs and a combination of 100 µM celecoxib and 240 µM fluvastatin was the most synergistic. This synergistic combination was used on the explant cultures and led to massive cell death of pilocytic astrocytoma cells.As a proof of concept, a patient with a refractory multifocal pilocytic astrocytoma was successfully treated with the fluvastatin/celecoxib combination used for 18 months. It was well tolerated and led to a partial tumor response. CONCLUSION: This study reports evidence for new targets and synergistic effect of celecoxib/fluvastatin combination in pilocytic astrocytoma. Because it is non-toxic, this new strategy offers hope for the treatment of patients with refractory pilocytic astrocytoma.


Subject(s)
Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Cyclooxygenase 2 Inhibitors/administration & dosage , Fatty Acids, Monounsaturated/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Indoles/administration & dosage , Pyrazoles/administration & dosage , Sulfonamides/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , Astrocytoma/pathology , Astrocytoma/physiopathology , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , CD36 Antigens/metabolism , Celecoxib , Cell Line, Tumor , Child, Preschool , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/adverse effects , Drug Synergism , Fatty Acids, Monounsaturated/adverse effects , Female , Fluvastatin , Gene Expression/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Indoles/adverse effects , Intercellular Adhesion Molecule-1/metabolism , Proto-Oncogene Proteins c-crk/metabolism , Pyrazoles/adverse effects , Sulfonamides/adverse effects , Tissue Culture Techniques , ras GTPase-Activating Proteins/metabolism
3.
Nat Genet ; 45(8): 927-32, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23817572

ABSTRACT

Pilocytic astrocytoma, the most common childhood brain tumor, is typically associated with mitogen-activated protein kinase (MAPK) pathway alterations. Surgically inaccessible midline tumors are therapeutically challenging, showing sustained tendency for progression and often becoming a chronic disease with substantial morbidities. Here we describe whole-genome sequencing of 96 pilocytic astrocytomas, with matched RNA sequencing (n = 73), conducted by the International Cancer Genome Consortium (ICGC) PedBrain Tumor Project. We identified recurrent activating mutations in FGFR1 and PTPN11 and new NTRK2 fusion genes in non-cerebellar tumors. New BRAF-activating changes were also observed. MAPK pathway alterations affected all tumors analyzed, with no other significant mutations identified, indicating that pilocytic astrocytoma is predominantly a single-pathway disease. Notably, we identified the same FGFR1 mutations in a subset of H3F3A-mutated pediatric glioblastoma with additional alterations in the NF1 gene. Our findings thus identify new potential therapeutic targets in distinct subsets of pilocytic astrocytoma and childhood glioblastoma.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Mutation , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, trkB/genetics , Animals , Astrocytoma/metabolism , Base Sequence , Brain Neoplasms/metabolism , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromosome Breakpoints , Chromosomes, Human, Pair 6 , Chromosomes, Human, Pair 9 , Fibroblast Growth Factors/metabolism , Humans , MAP Kinase Signaling System , Mice , Models, Molecular , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Protein Conformation , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, trkB/metabolism
4.
Acta Neuropathol ; 126(2): 291-301, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23660940

ABSTRACT

Pilocytic astrocytomas (PAs) are the most common brain tumors in pediatric patients and can cause significant morbidity, including chronic neurological deficiencies. They are characterized by activating alterations in the mitogen-activated protein kinase pathway, but little else is known about their development. To map the global DNA methylation profiles of these tumors, we analyzed 62 PAs and 7 normal cerebellum samples using Illumina 450K microarrays. These data revealed two subgroups of PA that separate according to tumor location (infratentorial versus supratentorial), and identified key neural developmental genes that are differentially methylated between the two groups, including NR2E1 and EN2. Integration with transcriptome microarray data highlighted significant expression differences, which were unexpectedly associated with a strong positive correlation between methylation and expression. Differentially methylated probes were often identified within the gene body and/or regions up- or downstream of the gene, rather than at the transcription start site. We also identified a large number of differentially methylated genes between cerebellar PAs and normal cerebellum, which were again enriched for developmental genes. In addition, we found a significant association between differentially methylated genes and SUZ12 binding sites, indicating potential disruption of the polycomb repressor complex 2 (PRC2). Taken together, these data suggest that PA from different locations in the brain may arise from region-specific cells of origin, and highlight the potential disruption of key developmental regulators during tumorigenesis. These findings have implications for future basic research and clinical trials, as therapeutic targets and drug sensitivity may differ according to tumor location.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Cerebellar Neoplasms/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Astrocytoma/pathology , Binding Sites/genetics , Brain Neoplasms/pathology , Cerebellar Neoplasms/pathology , Child , DNA Methylation/genetics , Gene Expression Profiling , Genes, Developmental/genetics , Humans , Neoplasm Proteins , Oligonucleotide Array Sequence Analysis , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Transcription Factors
5.
J Neurooncol ; 110(1): 21-5, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22814862

ABSTRACT

Rosette-forming glioneuronal tumors (RGNT) of the fourth ventricle are rare mixed glioneuronal tumors included in the revised WHO classification of central nervous system tumors, showing partial histological similarities to pilocytic astrocytomas. To evaluate potential similarities at the molecular level between these tumors, we analysed a series of 10 RGNT for the presence of KIAA1549-BRAF fusions using interphase fluorescence in situ hybridisation. However, we found no cases showing KIAA1549-BRAF gene fusion or BRAF (V600E) mutation. Our data support the hypothesis that RGNT may represent a distinct entity among the glioneuronal tumors of the central nervous system, with molecular features different from pilocytic astrocytomas.


Subject(s)
Brain Neoplasms/genetics , Fourth Ventricle/pathology , Ganglioglioma/genetics , Oncogene Fusion/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins B-raf/genetics , Adolescent , Adult , Brain Neoplasms/pathology , Cerebral Ventricle Neoplasms/genetics , Child , Female , Ganglioglioma/pathology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Rosette Formation
6.
J Neuropathol Exp Neurol ; 71(7): 631-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22710963

ABSTRACT

Cerebellar low-grade astrocytomas with a diffuse pattern of growth are uncommon, comprising World Health Organization (WHO) grade II diffuse astrocytomas (DA) and a minority of WHO grade I pilocytic astrocytomas (PA), so-called PA, "diffuse variant." Among 106 cerebellar low-grade astrocytomas (WHO grade I and II) operated on at the Mayo Clinic (1984-2010), we identified 19 such cases: 8 PA, "diffuse variant," 5 DA, and 6 that we were unable to classify further (low-grade astrocytomas, subtype indeterminate). We characterized these tumors using immunohistochemistry and currently available molecular markers (IDH1/2 mutations and BRAF mutation/fusion gene status) and investigated whether the markers could be used to aid the diagnostic process in combination with the clinical and pathologic features. KIAA1549-BRAF fusion was detected in 4 PA, "diffuse variant," 2 DA, and 2 low-grade astrocytomas, subtype indeterminate, indicating that these tumors were molecularly consistent with PA, the most common subtype of the series. A BRAF V600E mutation was detected in 1 PA, "diffuse variant" case; an IDH1 R132G mutation was found in 1 DA case. These results suggest that KIAA1549-BRAF fusion status and IDH1/2 and BRAF V600E mutational analyses may assist in the histologic classification of this diagnostically challenging group of tumors and result in a more accurate and objective combined molecular and histologic classification.


Subject(s)
Astrocytoma/metabolism , Cerebellar Neoplasms/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Adolescent , Adult , Astrocytoma/genetics , Astrocytoma/pathology , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Child , Child, Preschool , DNA Mutational Analysis , Female , Follow-Up Studies , Humans , Infant , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Male , Middle Aged , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Retrospective Studies , Young Adult
8.
Int J Cancer ; 131(3): E216-26, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22052591

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC) is the second most common form of nonmelanoma skin cancer (NMSC), and its incidence is increasing rapidly. Metastatic cSCC accounts for the majority of deaths associated with NMSC, but the genetic basis for cSCC progression remains poorly understood. A previous study identified small deletions (typically <1 Mb) in the protein tyrosine phosphatase receptor Type D (PTPRD) gene that segregated with more aggressive cSCC. To investigate the apparent association between deletion within PTPRD and cSCC metastasis, a series of 74 formalin-fixed paraffin-embedded tumors from 31 patients was analyzed using a custom Illumina 384 SNP microarray. Deletions were found in 37% of patients with metastatic cSCC and were strongly associated with metastatic tumors when compared to those that had not metastasized (p = 0.007). Subsequent mutation analysis revealed a higher mutation rate for PTPRD than has been reported in any other cancer type, with 37% of tumors harboring a somatic mutation. Conversely, bisulfite sequencing showed that methylation was not a mechanism of PTPRD disruption in cSCC. This is the first report to observe an association between deletion within PTPRD and metastatic disease and highlights the potential use of these deletions as a diagnostic biomarker for tumor progression. Combined with the high mutation rate observed in our study, PTPRD is one of the most commonly altered genes in cSCC and warrants further investigation to determine its significance for metastasis in other tumor types.


Subject(s)
Carcinoma, Squamous Cell/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Sequence Deletion , Skin Neoplasms/genetics , Base Sequence , Biomarkers, Tumor , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/secondary , Disease Progression , Genotype , Humans , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Paraffin Embedding , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Skin Neoplasms/pathology
9.
Biomaterials ; 32(33): 8538-47, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21824652

ABSTRACT

The CD95/CD95L receptor-ligand system is mainly recognised in the induction of apoptosis. However, it has also been shown that CD95L is over-expressed in many cancer types where it modulates immune-evasion and together with its receptor CD95 promotes tumour growth. Here, we show that CD95 surface modification of relatively large microparticles >0.5 µm in diameter, including those made from biodegradable polylactic-co-glycolic acid (PLGA), enhances intracellular uptake by a range of CD95L expressing cells in a process akin to phagocytosis. Using this approach we describe the intracellular uptake of microparticles and agent delivery in neurons, medulloblastoma, breast and ovarian cancer cells in vitro. CD95 modified paclitaxel-loaded PLGA microparticles are shown to be significantly more effective compared to conventional paclitaxel therapy (Taxol) at the same dose in subcutaneous medulloblastoma (∗∗∗P < 0.0001) and orthotopic ovarian cancer xenograft models where a >65-fold reduction in tumour bioluminescence was measured after treatment (∗P = 0.012). This drug delivery platform represents a new way of manipulating the normally advantageous tumour CD95L over-expression towards a therapeutic strategy. CD95 functionalised drug carriers could contribute to the improved function of cytotoxics in cancer, potentially increasing drug targeting and efficacy whilst reducing toxicity.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Lactic Acid , Microspheres , Paclitaxel/pharmacokinetics , Polyglycolic Acid , fas Receptor/chemistry , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Drug Carriers , Flow Cytometry , Humans , Paclitaxel/administration & dosage , Paclitaxel/chemistry , Phagocytosis , Polylactic Acid-Polyglycolic Acid Copolymer
10.
Clin Cancer Res ; 17(14): 4650-60, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21610151

ABSTRACT

PURPOSE: Oncogenic BRAF/Ras or NF1 loss can potentially trigger oncogene-induced senescence (OIS) through activation of the mitogen-activated protein kinase (MAPK) pathway. Somatic genetic abnormalities affecting this pathway occur in the majority of pilocytic astrocytomas (PA), the most prevalent brain neoplasm in children. We investigated whether OIS is induced in PA. EXPERIMENTAL DESIGN: We tested expression of established senescence markers in three independent cohorts of sporadic PA. We also assessed for OIS in vitro, using forced expression of wild-type and V600E-mutant BRAF in two astrocytic cell lines: human telomerase reverse transcriptase (hTERT)-immortalized astrocytes and fetal astrocytes. RESULTS: Our results indicate that PAs are senescent as evidenced by marked senescence-associated acidic ß-galactosidase activity, low KI-67 index, and induction of p16(INK4a) but not p53 in the majority of 52 PA samples (46 of 52; 88.5%). Overexpression of a number of senescence-associated genes [CDKN2A (p16), CDKN1A (p21), CEBPB, GADD45A, and IGFBP7] was shown at the mRNA level in two independent PA tumor series. In vitro, sustained activation of wild-type or mutant BRAF induced OIS in both astrocytic cell lines. Loss of p16(INK4a) in immortalized astrocytes abrogated OIS, indicative of the role of this pathway in mediating this phenomenon in astrocytes. OIS is a mechanism of tumor suppression that restricts the progression of benign tumors. We show that it is triggered in PAs through p16(INK4a) pathway induction following aberrant MAPK activation. CONCLUSIONS: OIS may account for the slow growth pattern in PA, the lack of progression to higher-grade astrocytomas, and the high overall survival of affected patients.


Subject(s)
Astrocytoma/genetics , Astrocytoma/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , MAP Kinase Signaling System/genetics , Oncogene Proteins/metabolism , Adolescent , Astrocytoma/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Cell Line , Cellular Senescence/genetics , Child , Child, Preschool , Cohort Studies , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Infant , Male , Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism
11.
Acta Neuropathol ; 121(6): 763-74, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21424530

ABSTRACT

Activation of the MAPK signaling pathway has been shown to be a unifying molecular feature in pilocytic astrocytoma (PA). Genetically, tandem duplications at chromosome 7q34 resulting in KIAA1549-BRAF fusion genes constitute the most common mechanism identified to date. To elucidate alternative mechanisms of aberrant MAPK activation in PA, we screened 125 primary tumors for RAF fusion genes and mutations in KRAS, NRAS, HRAS, PTPN11, BRAF and RAF1. Using microarray-based comparative genomic hybridization (aCGH), we identified in three cases an interstitial deletion of ~2.5 Mb as a novel recurrent mechanism forming BRAF gene fusions with FAM131B, a currently uncharacterized gene on chromosome 7q34. This deletion removes the BRAF N-terminal inhibitory domains, giving a constitutively active BRAF kinase. Functional characterization of the novel FAM131B-BRAF fusion demonstrated constitutive MEK phosphorylation potential and transforming activity in vitro. In addition, our study confirmed previously reported BRAF and RAF1 fusion variants in 72% (90/125) of PA. Mutations in BRAF (8/125), KRAS (2/125) and NF1 (4/125) and the rare RAF1 gene fusions (2/125) were mutually exclusive with BRAF rearrangements, with the exception of two cases in our series that concomitantly harbored more than one hit in the MAPK pathway. In summary, our findings further underline the fundamental role of RAF kinase fusion products as a tumor-specific marker and an ideally suited drug target for PA.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , MAP Kinase Signaling System/genetics , Mutation/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins B-raf/genetics , Animals , Child, Preschool , Chromosomes, Human, Pair 7/genetics , Comparative Genomic Hybridization , Female , Follow-Up Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Infant , Male , Mice , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Phosphorylation/genetics , Sequence Deletion , Statistics, Nonparametric , Transfection/methods
12.
Acta Neuropathol ; 121(6): 753-61, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21327941

ABSTRACT

Diffuse astrocytomas (WHO grade II) typically present as slow-growing tumours showing significant cellular differentiation, but possessing a tendency towards malignant progression. They account for ~10% of all astrocytic tumours, with a peak incidence between 30 and 40 years of age. Median survival is reported as around 6-8 years. Mutations of TP53 and IDH1 have been described as genetic hallmarks, while copy number alterations are also relatively common. However, there is some evidence to suggest that these characteristics may vary with age. Here, we present an integrated clinicopathologic, genomic and transcriptomic analysis suggesting that paediatric and adult tumours are associated with distinct genetic signatures. For example, no childhood tumour showed mutation of IDH1/2 or TP53, virtually no copy number changes were seen, and MGMT methylation was absent. In contrast, adult tumours showed IDH1/2 mutation in 94% and TP53 mutation in 69% of cases, with multiple copy number alterations per case and hypermethylation of MGMT in the majority of tumours. These differences were associated with a worse prognosis in the adult patients. The expression array data also revealed a significant difference in the expression of a number of genes putatively involved in neural stem cell maintenance and CNS development, including DLL3, HES5, BMP2, TIMP1 and BAMBI. Genes involved in DNA replication and the cell cycle were also enriched in the adult tumours, suggesting that their more aggressive behaviour may be due to derivation from a more rapidly dividing, less differentiated cell type.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Gene Expression Regulation, Neoplastic/genetics , Isocitrate Dehydrogenase/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Age Factors , Astrocytoma/pathology , Astrocytoma/physiopathology , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Child , DNA Copy Number Variations , DNA Methylation , DNA Mutational Analysis/methods , Female , Gene Expression Profiling/methods , Humans , Male , Middle Aged , Mutation/genetics , Oligonucleotide Array Sequence Analysis/methods , Principal Component Analysis , Survival Analysis , Young Adult
13.
J Invest Dermatol ; 129(6): 1562-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19131950

ABSTRACT

Cutaneous squamous cell carcinomas (cSCCs) are the second most frequent cancers in fair-skinned populations; yet, because of their genetic heterogeneity, the key molecular events in cSCC tumorigenesis remain poorly defined. We have used single nucleotide polymorphism microarray analysis to examine genome-wide allelic imbalance in 60 cSCCs using paired non-tumor samples. The most frequent recurrent aberrations were loss of heterozygosity at 3p and 9p, observed in 39 (65%) and 45 (75%) tumors, respectively. Microdeletions at 9p23 within the protein tyrosine phosphatase receptor type D (PTPRD) locus were identified in 9 (15%) samples, supporting a tumor suppressor role for PTPRD in cSCC. In addition, microdeletions at 3p14.2 were detected in 3 (5%) cSCCs, implicating the fragile histidine triad (FHIT) gene as a possible target for inactivation. Statistical analysis revealed that well-differentiated cSCCs demonstrated significantly fewer aberrations than moderately and poorly differentiated cSCCs; yet, despite a lower rate of allelic imbalance, some specific aberrations were observed equally frequently in both groups. No correlation was established between the frequency of chromosomal aberrations and immune or human papillomavirus status. Our data suggest that well-differentiated tumors are a genetically distinct subpopulation of cSCC.


Subject(s)
Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Oligonucleotide Array Sequence Analysis/methods , Polymorphism, Single Nucleotide , Skin Neoplasms/diagnosis , Skin Neoplasms/genetics , Acid Anhydride Hydrolases/genetics , Alleles , Chromosome Mapping , Cluster Analysis , Gene Deletion , Gene Expression Profiling , Genes, Tumor Suppressor , Humans , Loss of Heterozygosity , Models, Genetic , Neoplasm Proteins/genetics , Risk
14.
Eur J Trauma Emerg Surg ; 35(5): 470, 2009 Oct.
Article in English | MEDLINE | ID: mdl-26815213

ABSTRACT

INTRODUCTION: This study aims to determine, by outcome analysis, the appropriateness of current criteria employed to select patients for total hip arthroplasty (THA) as the primary treatment for displaced intracapsular hip fracture (DICHF). This study is intended to inform prospective randomised controlled trials investigating the efficacy of THA as a primary treatment. MATERIALS AND METHODS: Contemporary THA eligibility criteria were derived from recent publications relating to pre-fracture residence, mobility and independence. Outcome data were analysed for 96 patients (19% of 506 consecutive patients with DICHF between March 2003 and February 2005) who fulfilled those criteria. The variables analysed included age, gender, co-existing injuries, co-morbidities, social circumstances, mobility, independence, delay to surgery, readmission and death. The 96 patients included in the study were followed for 3 years. The primary outcome was the combined achievement of home or warden-assisted accommodation at 3 months, no patient readmission within 6 weeks of discharge and survival to 1 year. Secondary outcome was survival to 3 years. RESULTS: At 3 months, 86 patients (90%) had returned home, three (3.1%) required nursing or residential home placement, four (4.2%) were still resident in a community hospital and three (3.1%) had died. A total of ten patients failed to return to their own home. Eight patients (8.3%) were readmitted within 6 weeks. At 1 year, eight patients (8.3%) had died; by 3 years, the mortality rate was 25%. Patients not achieving return to home were older (84.8 years vs. 79.7 years, p = 0.19), prior to fracture weremore likely to use a walking aid (odds ratio [OR] 2.35) or required home support (OR 1.74). The number of co-morbidities was not an association. Backward selection applied to the above data produced an OR of 1.12 (confidence interval [CI] 1.01-1.21) for age as a variable in patients successfully discharged home, so that for each increase in age by year, the odds of home not being the final destination increased by 12%. These factors were also reflected in the patients requiring readmission within 6 weeks from discharge. CONCLUSIONS: If maintaining a high level of activity and independence is the expectation in patients considered for THA for DICHFs, then current selection criteria appear to be appropriate in identifying those capable of returning home, remaining independent and surviving to 1 year compared to the whole DICHF population. With a 75% 3-year survival, the postulated benefit of THA will not be realised in many patients and this needs to be considered. Cost-effectiveness trials are required before broad practice change occurs.

15.
Genes Chromosomes Cancer ; 46(7): 661-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17420988

ABSTRACT

Cutaneous squamous cell carcinomas (SCC) are the second most commonly diagnosed cancers in fair-skinned people; yet the genetic mechanisms involved in SCC tumorigenesis remain poorly understood. We have used single nucleotide polymorphism (SNP) microarray analysis to examine genome-wide allelic imbalance in 16 primary and 2 lymph node metastatic SCC using paired non-tumour samples to counteract normal copy number variation. The most common genetic change was loss of heterozygosity (LOH) on 9p, observed in 13 of 16 primary SCC. Other recurrent events included LOH on 3p (9 tumors), 2q, 8p, and 13 (each in 8 SCC) and allelic gain on 3q and 8q (each in 6 tumors). Copy number-neutral LOH was observed in a proportion of samples, implying that somatic recombination had led to acquired uniparental disomy, an event not previously demonstrated in SCC. As well as recurrent patterns of gross chromosomal changes, SNP microarray analysis revealed, in 2 primary SCC, a homozygous microdeletion on 9p23 within the protein tyrosine phosphatase receptor type D (PTPRD) locus, an emerging frequent target of homozygous deletion in lung cancer and neuroblastoma. A third sample was heterozygously deleted within this locus and PTPRD expression was aberrant. Two of the 3 primary SCC with PTPRD deletion had demonstrated metastatic potential. Our data identify PTPRD as a candidate tumor suppressor gene in cutaneous SCC with a possible association with metastasis.


Subject(s)
Alleles , Carcinoma, Squamous Cell/genetics , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatases/genetics , Skin Neoplasms/genetics , Aged , Aged, 80 and over , Chromosome Aberrations , Female , Humans , Loss of Heterozygosity , Male , Middle Aged , Receptor-Like Protein Tyrosine Phosphatases, Class 2
16.
Neonatal Netw ; 21(6): 15-21, 2002.
Article in English | MEDLINE | ID: mdl-12240510

ABSTRACT

PURPOSE: To establish the validity and clinical usefulness of a modified pain assessment scale, the Pain Assessment in Neonates (PAIN) scale. DESIGN: Correlational design to compare scores obtained on the PAIN with scores obtained on the Neonatal Infant Pain Scale (NIPS). SAMPLE: A convenience sample of 196 neonates from an NICU and a step-down unit with gestational ages of 26 to 47 weeks. METHOD: Bedside nurses observed the neonates for two minutes and then scored their responses on both scales. The scales were scored sequentially and in a randomized order. MAIN OUTCOME VARIABLE: Correlation of individual item scores and total scores on the PAIN and the NIPS. RESULTS: The scores for individual items on the PAIN were significantly associated with scores obtained on the NIPS. Overall correlation between the scales was 0.93. These associations suggest that the PAIN is a valid scale for assessment of neonatal pain.


Subject(s)
Pain Measurement/methods , Pain/diagnosis , Pain/nursing , Cohort Studies , Female , Humans , Infant, Newborn , Intensive Care Units, Neonatal , Male , Nursing Diagnosis , Pain/epidemiology , Pediatric Nursing/methods , Reproducibility of Results , Sampling Studies , Sensitivity and Specificity , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...