Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Clin Cancer Res ; 29(1): 261-270, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36260525

ABSTRACT

PURPOSE: Chordoma is a rare bone tumor with a high recurrence rate and limited treatment options. The aim of this study was to identify molecular subtypes of chordoma that may improve clinical management. EXPERIMENTAL DESIGN: We conducted RNA sequencing in 48 tumors from patients with Chinese skull-base chordoma and identified two major molecular subtypes. We then replicated the classification using a NanoString panel in 48 patients with chordoma from North America. RESULTS: Tumors in one subtype were more likely to have somatic mutations and reduced expression in chromatin remodeling genes, such as PBRM1 and SETD2, whereas the other subtype was characterized by the upregulation of genes in epithelial-mesenchymal transition and Sonic Hedgehog pathways. IHC staining of top differentially expressed genes between the two subtypes in 312 patients with Chinese chordoma with long-term follow-up data showed that the expression of some markers such as PTCH1 was significantly associated with survival outcomes. CONCLUSIONS: Our findings may improve the understanding of subtype-specific tumorigenesis of chordoma and inform clinical prognostication and targeted options.


Subject(s)
Chordoma , Skull Base Neoplasms , Humans , Chordoma/genetics , Chordoma/pathology , Hedgehog Proteins/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Profiling , Skull Base Neoplasms/genetics , Skull Base Neoplasms/pathology
2.
Hum Mutat ; 43(10): 1396-1407, 2022 10.
Article in English | MEDLINE | ID: mdl-35762214

ABSTRACT

Chordoma is a rare bone tumor with genetic risk factors largely unknown. We conducted a whole-exome sequencing (WES) analysis of germline DNA from 19 familial chordoma cases in five pedigrees and 137 sporadic chordoma patients and identified 17 rare germline variants in PALB2 and BRCA2, whose products play essential roles in homologous recombination (HR) and tumor suppression. One PALB2 variant showed disease cosegregation in a family with four affected people or obligate gene carrier. Chordoma cases had a significantly increased burden of rare variants in both genes when compared to population-based controls. Four of the six PALB2 variants identified from chordoma patients modestly affected HR function and three of the 11 BRCA2 variants caused loss of function in experimental assays. These results, together with previous reports of abnormal morphology and Brachyury expression of the notochord in Palb2 knockout mouse embryos and genomic signatures associated with HR defect and HR gene mutations in advanced chordomas, suggest that germline mutations in PALB2 and BRCA2 may increase chordoma susceptibility. Our data shed light on the etiology of chordoma and support the previous finding that PARP-1 inhibitors may be a potential therapy for some chordoma patients.


Subject(s)
BRCA2 Protein , Breast Neoplasms , Chordoma , Fanconi Anemia Complementation Group N Protein , Animals , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Chordoma/genetics , Fanconi Anemia Complementation Group N Protein/genetics , Female , Genes, BRCA2 , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Mice
3.
Cancers (Basel) ; 13(11)2021 May 30.
Article in English | MEDLINE | ID: mdl-34070849

ABSTRACT

BACKGROUND: Chordoma is a rare bone cancer with an unknown etiology. TBXT is the only chordoma susceptibility gene identified to date; germline single nucleotide variants and copy number variants in TBXT have been associated with chordoma susceptibility in familial and sporadic chordoma. However, the genetic susceptibility of chordoma remains largely unknown. In this study, we investigated rare germline genetic variants in genes involved in TBXT/chordoma-related signaling pathways and other biological processes in chordoma patients from North America and China. METHODS: We identified variants that were very rare in general population and internal control datasets and showed evidence for pathogenicity in 265 genes in a whole exome sequencing (WES) dataset of 138 chordoma patients of European ancestry and in a whole genome sequencing (WGS) dataset of 80 Chinese patients with skull base chordoma. RESULTS: Rare and likely pathogenic variants were identified in 32 of 138 European ancestry patients (23%), including genes that are part of notochord development, PI3K/AKT/mTOR, Sonic Hedgehog, SWI/SNF complex and mesoderm development pathways. Rare pathogenic variants in COL2A1, EXT1, PDK1, LRP2, TBXT and TSC2, among others, were also observed in Chinese patients. CONCLUSION: We identified several rare loss-of-function and predicted deleterious missense variants in germline DNA from patients with chordoma, which may influence chordoma predisposition and reflect a complex susceptibility, warranting further investigation in large studies.

4.
Nat Commun ; 12(1): 757, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536423

ABSTRACT

Chordoma is a rare bone tumor with an unknown etiology and high recurrence rate. Here we conduct whole genome sequencing of 80 skull-base chordomas and identify PBRM1, a SWI/SNF (SWItch/Sucrose Non-Fermentable) complex subunit gene, as a significantly mutated driver gene. Genomic alterations in PBRM1 (12.5%) and homozygous deletions of the CDKN2A/2B locus are the most prevalent events. The combination of PBRM1 alterations and the chromosome 22q deletion, which involves another SWI/SNF gene (SMARCB1), shows strong associations with poor chordoma-specific survival (Hazard ratio [HR] = 10.55, 95% confidence interval [CI] = 2.81-39.64, p = 0.001) and recurrence-free survival (HR = 4.30, 95% CI = 2.34-7.91, p = 2.77 × 10-6). Despite the low mutation rate, extensive somatic copy number alterations frequently occur, most of which are clonal and showed highly concordant profiles between paired primary and recurrence/metastasis samples, indicating their importance in chordoma initiation. In this work, our findings provide important biological and clinical insights into skull-base chordoma.


Subject(s)
Chordoma/genetics , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease/genetics , SMARCB1 Protein/genetics , Skull Base Neoplasms/genetics , Transcription Factors/genetics , Whole Genome Sequencing/methods , Adult , Chordoma/pathology , DNA Copy Number Variations , Female , Genomics/methods , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Mutation , Neoplasm Recurrence, Local , Skull Base Neoplasms/pathology , Young Adult
5.
J Neurosurg ; 134(5): 1399-1408, 2020 Jun 19.
Article in English | MEDLINE | ID: mdl-32559743

ABSTRACT

OBJECTIVE: To gain insight into the role of germline genetics in the development of chordoma, the authors evaluated data from 2 sets of patients with familial chordoma, those with and without a germline duplication of the T gene (T-dup+ vs T-dup-), which was previously identified as a susceptibility mechanism in some families. The authors then compared the patients with familial tumors to patients with sporadic chordoma in the US general population reported to the National Cancer Institute's Surveillance, Epidemiology, and End Results (SEER) program through 2015. METHODS: Evaluation of family members included review of personal and family medical history, physical and neurological examination, and pre- and postcontrast MRI of the skull base and spine. Sixteen patients from 6 white families with chordoma had a chordoma diagnosis at family referral. Screening MR images of 35 relatives revealed clival lesions in 6, 4 of which were excised and confirmed to be chordoma. Thus, data were available for 20 patients with histologically confirmed familial chordoma. There were 1759 patients with histologically confirmed chordoma in SEER whose race was known. RESULTS: The median age at chordoma diagnosis differed across the groups: it was lowest in T-dup+ familial patients (26.8 years, range 5.3-68.4 years); intermediate in T-dup- patients (46.2 years, range 11.8-60.1 years); and highest in SEER patients (57 years, range 0-98 years). There was a marked preponderance of skull base tumors in patients with familial chordoma (93% in T-dup+ and 83% in T-dup-) versus 38% in the SEER program (37% in white, 53% in black, and 48.5% in Asian/Pacific Islander/American Indian/Alaska Native patients). Furthermore, 29% of white and 16%-17% of nonwhite SEER patients had mobile-spine chordoma, versus no patients in the familial group. Several T-dup+ familial chordoma patients had putative second/multiple primary chordomas. CONCLUSIONS: The occurrence of young age at diagnosis, skull base presentation, or multiple primary chordomas should encourage careful review of family history for patients diagnosed with chordoma as well as screening of at-risk family members by MRI for early detection of chordoma. Furthermore, given genetic predisposition in some patients with familial chordoma, identification of a specific mutation in a family will permit surveillance to be limited to mutation carriers-and consideration should be given for imaging the entire neuraxis in any chordoma patient presenting at an early age or with a blood relative with chordoma. Finally, future studies should explore racial differences in age at diagnosis and presenting site in chordoma.


Subject(s)
Chordoma/genetics , Neoplastic Syndromes, Hereditary/genetics , Skull Base Neoplasms/genetics , Spinal Neoplasms/genetics , T-Box Domain Proteins/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Chordoma/epidemiology , Chordoma/pathology , Coccyx , Ethnicity/genetics , Female , Gene Duplication , Genetic Testing , Germ-Line Mutation , Humans , Infant , Male , Middle Aged , Neoplastic Syndromes, Hereditary/epidemiology , Neoplastic Syndromes, Hereditary/pathology , Pedigree , SEER Program , Sacrum , Skull Base Neoplasms/epidemiology , Skull Base Neoplasms/pathology , Spinal Neoplasms/epidemiology , Spinal Neoplasms/pathology , United States/epidemiology , Young Adult
6.
Hum Genet ; 133(10): 1289-97, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24990759

ABSTRACT

Chordoma is a rare bone cancer that is believed to originate from notochordal remnants. We previously identified germline T duplication as a major susceptibility mechanism in several chordoma families. Recently, a common genetic variant in T (rs2305089) was significantly associated with the risk of sporadic chordoma. We sequenced all T exons in 24 familial cases and 54 unaffected family members from eight chordoma families (three with T duplications), 103 sporadic cases, and 160 unrelated controls. We also measured T copy number variation in all sporadic cases. We confirmed the association between the previously reported variant rs2305089 and risk of familial [odds ratio (OR) = 2.6, 95% confidence interval (CI) = 0.93, 7.25, P = 0.067] and sporadic chordoma (OR = 2.85, 95% CI = 1.89, 4.29, P < 0.0001). We also identified a second common variant, rs1056048, that was strongly associated with chordoma in families (OR = 4.14, 95% CI = 1.43, 11.92, P = 0.0086). Among sporadic cases, another common variant (rs3816300) was significantly associated with risk when jointly analyzed with rs2305089. The association with rs3816300 was significantly stronger in cases with early age onset. In addition, we identified three rare variants that were only observed among sporadic chordoma cases, all of which have potential functional relevance based on in silico predictions. Finally, we did not observe T duplication in any sporadic chordoma case. Our findings further highlight the importance of the T gene in the pathogenesis of both familial and sporadic chordoma and suggest a complex susceptibility related to T.


Subject(s)
Chordoma/genetics , Fetal Proteins/genetics , Gene Duplication , Germ-Line Mutation , Skull Base Neoplasms/genetics , Spinal Neoplasms/genetics , T-Box Domain Proteins/genetics , Adolescent , Adult , Child , DNA Mutational Analysis , Family , Female , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Sacrum
7.
J Med Genet ; 48(7): 444-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21266383

ABSTRACT

BACKGROUND: Chordoma, an age-dependent rare cancer, arises from notochordal remnants. Fewer than 5% of chordomas occur in children. Tuberous sclerosis complex (TSC) is an autosomal dominant neurocutaneous syndrome characterised by abnormal tissue growths in multiple organ systems. Reports of chordoma in children with TSC suggest that TSC1 and TSC2 mutations may contribute to chordoma aetiology. METHODS: To determine whether the 10 TSC-associated childhood chordomas reported in the literature are representative of chordoma in the general paediatric population, the authors compared age at diagnosis, primary site and outcome in them with results from a systematic assessment of 65 paediatric chordoma cases reported to the US population-based cancer registries contributing to the SEER Program of the National Cancer Institute. RESULTS: TSC-associated paediatric chordomas differed from chordomas in the general paediatric population: median age at diagnosis (6.2 months, TSC, vs 12.5 years, SEER); anatomical site (40% sacral, TSC, vs 9.4% sacral, SEER); and site-specific age at diagnosis (all four sacral chordomas diagnosed during the fetal or neonatal period, TSC, vs all six sacral chordomas diagnosed at >15 years, SEER). Finally, three of four patients with TSC-associated sacral chordoma were alive and tumour-free at 2.2, 8 and 19 years after diagnosis versus a median survival of 36 months among paediatric patients with sacral chordoma in SEER. CONCLUSIONS: These results strengthen the association between paediatric chordoma and TSC. Future clinical and molecular studies documenting the magnitude and clinical spectrum of the joint occurrence of these two diseases should provide the basis for delineating the biological relationship between them.


Subject(s)
Chordoma/diagnosis , Chordoma/etiology , Tuberous Sclerosis/complications , Tuberous Sclerosis/diagnosis , Adolescent , Age Factors , Child , Child, Preschool , Chordoma/genetics , Chordoma/mortality , Female , Humans , Infant , Infant, Newborn , Kaplan-Meier Estimate , Male , Mutation/genetics , Prognosis , SEER Program , Tuberous Sclerosis/genetics , Tuberous Sclerosis/mortality , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
8.
Am J Med Genet A ; 152A(3): 601-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20186797

ABSTRACT

We present monozygotic twins discordant for the autosomal dominant disorder neurofibromatosis type 1 (NF1). The affected twin was diagnosed with NF1 at age 12, based upon accepted clinical criteria for the disorder. Both twins were re-examined at ages 35 and 57, at which times the unaffected twin continued to show no clinical manifestations of NF1. Short tandem repeat marker (STR) genotyping at 10 loci on chromosome 17 and 10 additional loci dispersed across the genome revealed identical genotypes for the twins, confirming their monozygosity. The affected twin has three children, two of whom also have NF1, while the unaffected twin has two children, both unaffected. Using lymphoblastoid, fibroblast, and buccal cell samples collected from both twins and from other family members in three generations, we discovered a pathogenic nonsense mutation in exon 40 of the NF1 gene. This mutation was found in all cell samples from the affected twin and her affected daughter, and in lymphoblastoid and buccal cells but not fibroblasts from the unaffected twin. We also found a novel non-synonymous change in exon 16 of the NF1 gene that was transmitted from the unaffected mother to both twins and co-segregated with the pathogenic mutation in the ensuing generation. All cells from the twins were heterozygous for this apparent exon 16 polymorphism and for single nucleotide polymorphisms (SNPs) within 2.5 kb flanking the site of the exon 40 nonsense mutation. This suggests that the NF1 gene of the unaffected twin differed in the respective lymphoblastoid cells and fibroblasts only at the mutation site itself, making post-zygotic mutation leading to mosaicism the most likely mechanism of phenotypic discordance. Although the unaffected twin is a mosaic, the distribution of the mutant allele among different cells and tissues appears to be insufficient to cause overt clinical manifestations of NF1.


Subject(s)
Codon, Nonsense , Diseases in Twins/genetics , Genes, Neurofibromatosis 1 , Neurofibromatosis 1/genetics , Adult , Base Sequence , Child , DNA Mutational Analysis , Exons , Female , Heterozygote , Humans , Male , Middle Aged , Mosaicism , Pedigree , Polymorphism, Single Nucleotide , Twins, Monozygotic/genetics
9.
Nat Genet ; 41(11): 1176-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19801981

ABSTRACT

Using high-resolution array-CGH, we identified unique duplications of a region on 6q27 in four multiplex families with at least three cases of chordoma, a cancer of presumed notochordal origin. The duplicated region contains only the T (brachyury) gene, which is important in notochord development and is expressed in most sporadic chordomas. Our findings highlight the value of screening for complex genomic rearrangements in searches for cancer-susceptibility genes.


Subject(s)
Chordoma/genetics , Fetal Proteins/genetics , Gene Duplication , Genetic Predisposition to Disease , T-Box Domain Proteins/genetics , Chromosomes, Human, Pair 6 , Comparative Genomic Hybridization , Genome-Wide Association Study , Humans , Polymorphism, Single Nucleotide
10.
Lancet ; 373(9679): 1974-86, 2009 Jun 06.
Article in English | MEDLINE | ID: mdl-19476995

ABSTRACT

Neurofibromatosis type 2 is an autosomal-dominant multiple neoplasia syndrome that results from mutations in the NF2 tumour suppressor gene located on chromosome 22q. It has a frequency of one in 25,000 livebirths and nearly 100% penetrance by 60 years of age. Half of patients inherit a germline mutation from an affected parent and the remainder acquire a de novo mutation for neurofibromatosis type 2. Patients develop nervous system tumours (schwannomas, meningiomas, ependymomas, astrocytomas, and neurofibromas), peripheral neuropathy, ophthalmological lesions (cataracts, epiretinal membranes, and retinal hamartomas), and cutaneous lesions (skin tumours). Optimum treatment is multidisciplinary because of the complexities associated with management of the multiple, progressive, and protean lesions associated with the disorder. We review the molecular pathogenesis, genetics, clinical findings, and management strategies for neurofibromatosis type 2.


Subject(s)
Neurofibromatosis 2 , Cataract/etiology , Chromosomes, Human, Pair 22/genetics , Disease Progression , Gene Frequency/genetics , Genes, Neurofibromatosis 2 , Genetic Testing , Humans , Molecular Biology , Mutation/genetics , Nervous System Neoplasms/etiology , Neurofibromatosis 2/diagnosis , Neurofibromatosis 2/epidemiology , Neurofibromatosis 2/genetics , Neurofibromatosis 2/therapy , Neurofibromin 2/genetics , Patient Care Team/organization & administration , Pedigree , Penetrance , Peripheral Nervous System Diseases/etiology , Skin Neoplasms/etiology , Survival Rate
11.
Eur J Neurosci ; 26(4): 943-52, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17714188

ABSTRACT

The periaqueductal grey can differentially control A- vs. C-nociceptor-evoked spinal reflexes and deep spinal dorsal horn neuronal responses. However, little is known about the control of A- vs. C-fibre inputs to lamina I and the lateral spinal nucleus, and how this correlates with the control of deeper laminae. To address this, the laminar distributions of neurones expressing Fos-like immunoreactivity were determined following preferential activation of A- or C-heat nociceptors, using fast or slow rates of skin heating, respectively, in the absence or presence of descending control evoked from the periaqueductal grey. In lamina I, numbers of Fos-positive neurones following both fast and slow rates of skin heating were reduced significantly following activation in the ventrolateral and dorsolateral/lateral periaqueductal grey. In contrast, in the deep dorsal horn (laminae III-VI), activation in both the ventrolateral and dorsolateral/lateral periaqueductal grey significantly reduced the numbers of Fos-positive neurones evoked by C- but not A-nociceptor stimulation. C- but not A-heat nociceptor activation evoked Fos bilaterally in the lateral spinal nucleus. Stimulation in the ventrolateral but not the dorsolateral/lateral periaqueductal grey significantly increased the numbers of Fos-positive neurones evoked by A- and C-nociceptor stimulation bilaterally in the lateral spinal nucleus. These data have demonstrated differences in the descending control of the superficial vs. the deep dorsal horn and lateral spinal nucleus with respect to the processing of A- and C-fibre-evoked events. The data are discussed in relation to the roles of A- and C-nociceptors in acute and chronic pain.


Subject(s)
Nerve Fibers, Myelinated/physiology , Nerve Fibers, Unmyelinated/physiology , Nociceptors/physiology , Periaqueductal Gray/physiology , Posterior Horn Cells/physiology , Animals , Cell Count , Data Interpretation, Statistical , Electric Stimulation , Homocysteine/analogs & derivatives , Homocysteine/pharmacology , Hot Temperature , Male , Microinjections , Neural Pathways/physiology , Peripheral Nerves/physiology , Proto-Oncogene Proteins c-fos/physiology , Rats , Rats, Wistar
12.
J Neurosurg Spine ; 5(2): 179; author reply 179, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16925089
13.
Int J Cancer ; 116(3): 487-91, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-15818627

ABSTRACT

Chordoma, a rare bone tumor originating from notochordal remnants, has a genetic predisposition in some families. Previously, we performed linkage analysis using microsatellite (STR) markers on 3 unrelated chordoma kindreds (16 patients with chordoma) and reported significant evidence for linkage to chromosome 7q33 (Z(max) = 4.78) with a minimal disease gene region of 11 cM. In our present study, we performed linkage analysis in these 3 families using chromosome 7 single nucleotide polymorphisms (SNPs). Parametric and nonparametric multipoint analyses showed significant linkage to 7q markers with p < 0.001 and Z(max) = 2.77, respectively. The minimal disease gene region was not reduced by combined SNP and STR haplotype analysis compared to the previous STR haplotype analysis alone. We genotyped members of a fourth chordoma family with SNP and STR markers for chromosome 7q and for 1p36, the location of a previously reported chordoma locus. Affected members of this family did not share a common haplotype on 7q, and the family did not show evidence of linkage to 1p36. Thus, we corroborated a chordoma locus on chromosome 7q in the 3 original families and demonstrated evidence for genetic heterogeneity in the fourth family. Our study also provided insights into some limitations and analytical complexities associated with using a dense SNP marker set in linkage analysis of complex pedigrees.


Subject(s)
Chordoma/genetics , Chromosomes, Human, Pair 7 , Genetic Markers , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Adult , Child , Female , Genetic Linkage , Genotype , Humans , Male , Middle Aged , Pedigree
14.
J Pathol ; 198(1): 14-20, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12210058

ABSTRACT

Individuals affected with the neurofibromatosis 2 (NF2) cancer predisposition syndrome develop specific ocular lesions. To determine whether these lesions result from altered NF2 gene expression, microdissection and PCR were used to investigate 40 ocular lesions from seven eyes of four NF2 patients for LOH, with markers that flank the NF2 gene on chromosome 22q. NF2 protein (merlin) expression was also evaluated in these lesions, using immunohistochemistry. Retinal hamartoma was observed in all seven eyes, including one with combined pigment epithelial and retinal hamartoma (CPERH). Retinal tufts were present in four eyes (three patients), retinal dysplasia in two eyes (two patients), optic nerve neurofibroma in one eye, iris naevoid hyperplasia in two eyes (two patients) and pseudophakia in all eyes. Markers were informative in three patients (six eyes from three unrelated families). One patient was non-informative due to prolonged decalcification. All retinal and optic nerve, but not iris lesions, demonstrated consistent LOH for the NF2 gene. Merlin was not expressed in the retina, optic nerve, or iris lesions. These results suggest that inactivation of the NF2 gene is associated with the formation of a variety of retinal and optic nerve lesions in NF2 patients.


Subject(s)
Genes, Neurofibromatosis 2 , Loss of Heterozygosity , Neurofibromatosis 2/genetics , Optic Nerve Neoplasms/genetics , Retinal Diseases/genetics , Adolescent , Adult , Aged , Chromosomes, Human, Pair 22/genetics , Hamartoma/genetics , Hamartoma/metabolism , Hamartoma/pathology , Humans , Male , Middle Aged , Neoplasm Proteins/metabolism , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Neurofibromin 2/metabolism , Optic Nerve Neoplasms/metabolism , Optic Nerve Neoplasms/pathology , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Dysplasia/genetics , Retinal Dysplasia/metabolism , Retinal Dysplasia/pathology
15.
J Neurosurg ; 96(2): 217-22, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11838793

ABSTRACT

OBJECT: The results of two longitudinal studies of growth rates of vestibular schwannomas (VSs) in patients with neurofibromatosis Type 2 (NF2) differ as to whether VS growth rates decrease or increase with increasing patient age. The authors undertook this study to assess the relationship between VS growth rates and patient age and type of constitutional NF2 mutation; they also examined variability in VS growth rates among multiple patients in families with NF2. METHODS: Gadolinium-enhanced magnetic resonance images obtained in 18 patients with inherited NF2 from 11 unrelated families were retrospectively analyzed. The patients had been observed for a median of 4 years. Volumes of the VSs were measured using a two-component box model (intrameatal and extrameatal parts measured separately). Single-strand conformation polymorphism analysis and Southern blot analysis were used to identify constitutional NF2 mutations. Growth rates of the VSs were highly variable, but tended to decrease with increasing patient age both at onset of signs or symptoms of NF2 (r2 = 0.35, p = 0.026) and at diagnosis (r2 = 0.33, p = 0.012). The VS growth rates did not vary significantly with the type of constitutional NF2 mutation or the number of non-VS cerebral or spinal tumors. The VS growth rates were highly variable within families and did not correspond to clinical indices of NF2 disease severity, such as patient age at symptom onset and the number of non-VS cerebral and spinal tumors. CONCLUSIONS: The growth rates of VSs in patients with NF2 are highly variable, but tend to decrease with increasing patient age. Clinical treatment of multiple patients in families with NF2 cannot be based on the expectations of similar VS growth rates, even when other clinical aspects of disease severity are similar.


Subject(s)
Neurofibromatosis 2/pathology , Neurofibromatosis 2/physiopathology , Neuroma, Acoustic/pathology , Neuroma, Acoustic/physiopathology , Adolescent , Adult , Age Factors , Aged , Blotting, Southern , Brain/pathology , Brain/physiopathology , Child , Child, Preschool , Female , Gadolinium , Genes, Neurofibromatosis 2/physiology , Humans , Longitudinal Studies , Magnetic Resonance Imaging , Male , Middle Aged , Mutation/genetics , Neurofibromatosis 2/genetics , Neuroma, Acoustic/genetics , Polymorphism, Single-Stranded Conformational , Predictive Value of Tests , Retrospective Studies , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...