77 results on '"Miriam J. Smith"'
Search Results
2. Dominant‐negative pathogenic variant <scp>BRIP1</scp> c. <scp>1045G</scp> >C is a high‐risk allele for non‐mucinous epithelial ovarian cancer: A case‐control study
- Author
-
Stephanie Amico, Nicola Flaum, Olivia Smith, Emma J Crosbie, Richard J. Edmondson, D. Gareth Evans, Elke M van Veen, William G. Newman, and Miriam J. Smith
- Subjects
Oncology ,medicine.medical_specialty ,endocrine system diseases ,Carcinoma, Ovarian Epithelial ,symbols.namesake ,Breast cancer ,Gene Frequency ,Internal medicine ,Genetics ,medicine ,Humans ,Missense mutation ,Genetic Predisposition to Disease ,Epithelial ovarian cancer ,Family history ,Gene ,Alleles ,Genetic Association Studies ,Genetics (clinical) ,Aged ,Genes, Dominant ,Ovarian Neoplasms ,Sanger sequencing ,Manchester Cancer Research Centre ,business.industry ,ResearchInstitutes_Networks_Beacons/mcrc ,BRIP1 ,Case-control study ,Sequence Analysis, DNA ,Middle Aged ,medicine.disease ,Fanconi Anemia Complementation Group Proteins ,female genital diseases and pregnancy complications ,Case-Control Studies ,symbols ,Female ,business ,RNA Helicases - Abstract
BRIP1 is a moderate susceptibility epithelial ovarian cancer (EOC) gene. Having identified the BRIP1 c.1045G>C missense variant in a number of families with EOC, we aimed to investigate the frequency of this and BRIP1.2392C>T pathogenic variant in patients with breast cancer (BC) and/or EOC. A case-control study of 3,767 cases and 2,043 controls was undertaken investigating the presence of these variants using Sanger sequencing and gene panel data. Individuals with BC and/or EOC were grouped by family history. BRIP1 c.1045G>C was associated with increased risk of BC/EOC (OR = 37.7; 95% CI 5.3-444.2; P=0.0001). The risk was highest for women with EOC (OR=140.8; 95% CI 23.5-1723.0; PT was associated with smaller risks for BC/EOC (OR=5.4; 95%CI 2.4-12.7; P=0.0003), EOC (OR=5.9; 95% CI 1.3-23.0; p=0.0550), and BC (OR=5.3; 95%CI 2.3-12.9; P=0.0009). Our study highlights the importance of BRIP1 as an EOC susceptibility gene, especially in familial EOC. The variant BRIP1 c.1045G>C, rs149364097, is of particular interest as its dominant-negative effect may confer a higher risk of EOC than that of the previously reported BRIP1 c.2392C>T nonsense variant. Dominant-negative missense variants may confer higher risks than their loss-of-function counterparts.
- Published
- 2021
3. Clinical utility of testing for PALB2 and CHEK2 c.1100delC in breast and ovarian cancer
- Author
-
Elaine F. Harkness, Sacha J Howell, Fiona Lalloo, Jamie M Ellingford, Miriam J. Smith, Anthony Howell, D. Gareth Evans, Helene Schlech, William G. Newman, George J Burghel, Claire Forde, Helen Byers, Naomi L. Bowers, Elke M van Veen, Emma R. Woodward, and Andrew J Wallace
- Subjects
0301 basic medicine ,medicine.medical_specialty ,PALB2 ,Breast Neoplasms ,Gastroenterology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,medicine ,Odds Ratio ,Humans ,In patient ,Genetic Predisposition to Disease ,skin and connective tissue diseases ,CHEK2 ,Triple negative ,Genetics (clinical) ,Ovarian Neoplasms ,business.industry ,Carcinoma in situ ,Odds ratio ,medicine.disease ,Checkpoint Kinase 2 ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Ovarian cancer ,business ,Fanconi Anemia Complementation Group N Protein - Abstract
PURPOSE To investigate the contribution of PALB2 pathogenic gene variants (PGVs, PALB2_PGV) and the CHEK2 c.1100delC (CHEK2_1100delC) PGV to familial breast and ovarian cancer, and PALB2_PGV associated breast cancer pathology. METHODS Outcomes of germline PALB2_PGV and CHEK2_1100delC testing were recorded in 3,127 women with histologically confirmed diagnoses of invasive breast cancer, carcinoma in situ, or epithelial nonmucinous ovarian cancer, and 1,567 female controls. Breast cancer pathology was recorded in PALB2_PGV cases from extended families. RESULTS Thirty-five PALB2 and 44 CHEK2_1100delC PGVs were detected in patients (odds ratio [OR] PALB2 breast-ovarian = 5.90 [95% CI: 1.92-18.36], CHEK2 breast-ovarian = 4.46 [95% CI: 1.86-10.46], PALB2 breast = 6.16 [95% CI: 1.98-19.21], CHEK2 breast = 4.89 [95% CI: 2.01-11.34]). Grade 3 ER-positive HER2-negative, grade 3 and triple negative (TN) tumors were enriched in cases with PALB2 PGVs compared with all breast cancers known to our service (respectively: 15/43, 254/1,843, P = 0.0005; 28/37, 562/1,381, P = 0.0001; 12/43, 204/1,639, P
- Published
- 2021
4. Current recommendations for cancer surveillance in Gorlin syndrome: a report from the SIOPE host genome working group (SIOPE HGWG)
- Author
-
O Michaeli, N Waespe, Laurence Brugières, Christian P. Kratz, Miriam J. Smith, Alexandra Russo, Steffen Hirsch, D G Evans, Saskia M. J. Hopman, B Doergeloh, H Salvador, V. Ridola, M. Jorgensen, T Milde, Léa Guerrini-Rousseau, B Claret, and M Kuhlen
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,Skin Neoplasms ,SUFU ,medicine.medical_treatment ,030105 genetics & heredity ,Gorlin syndrome ,0302 clinical medicine ,Epidemiology ,Genotype ,PTCH1 ,Keratocyst ,610 Medicine & health ,Child ,Genetics (clinical) ,Surveillance ,Basal Cell Nevus Syndrome ,Cancer predisposition syndrome ,Patched-1 Receptor ,Hereditary ,030220 oncology & carcinogenesis ,Child, Preschool ,Original Article ,medicine.symptom ,360 Social problems & social services ,medicine.medical_specialty ,Host genome ,03 medical and health sciences ,Young Adult ,Internal medicine ,Genetics ,medicine ,Humans ,Hedgehog Proteins ,ddc:610 ,Cerebellar Neoplasms ,business.industry ,Cancer ,medicine.disease ,Human genetics ,Radiation therapy ,Repressor Proteins ,stomatognathic diseases ,business - Abstract
Gorlin syndrome (MIM 109,400), a cancer predisposition syndrome related to a constitutional pathogenic variation (PV) of a gene in the Sonic Hedgehog pathway (PTCH1 or SUFU), is associated with a broad spectrum of benign and malignant tumors. Basal cell carcinomas (BCC), odontogenic keratocysts and medulloblastomas are the main tumor types encountered, but meningiomas, ovarian or cardiac fibromas and sarcomas have also been described. The clinical features and tumor risks are different depending on the causative gene. Due to the rarity of this condition, there is little data on phenotype-genotype correlations. This report summarizes genotype-based recommendations for screening patients with PTCH1 and SUFU-related Gorlin syndrome, discussed during a workshop of the Host Genome Working Group of the European branch of the International Society of Pediatric Oncology (SIOPE HGWG) held in January 2020. In order to allow early detection of BCC, dermatologic examination should start at age 10 in PTCH1, and at age 20 in SUFU PV carriers. Odontogenic keratocyst screening, based on odontologic examination, should begin at age 2 with annual orthopantogram beginning around age 8 for PTCH1 PV carriers only. For medulloblastomas, repeated brain MRI from birth to 5 years should be proposed for SUFU PV carriers only. Brain MRI for meningiomas and pelvic ultrasound for ovarian fibromas should be offered to both PTCH1 and SUFU PV carriers. Follow-up of patients treated with radiotherapy should be prolonged and thorough because of the risk of secondary malignancies. Prospective evaluation of evidence of the effectiveness of these surveillance recommendations is required.
- Published
- 2021
5. High likelihood of actionable pathogenic variant detection in breast cancer genes in women with very early onset breast cancer
- Author
-
Stephanie L Greville-Haygate, Emma R. Woodward, D. Gareth Evans, Andrew J Wallace, Helen Byers, Jamie M Ellingford, Fiona Lalloo, Anthony Howell, George J Burghel, Sasha J Howell, Miriam J. Smith, Elaine F. Harkness, William G. Newman, Naomi L. Bowers, Elke M van Veen, Diana Eccles, Sarah J Evans, and Marta Pereira
- Subjects
0301 basic medicine ,Oncology ,Adult ,medicine.medical_specialty ,endocrine system diseases ,PALB2 ,Genes, BRCA2 ,Genes, BRCA1 ,human genetics ,Breast Neoplasms ,Disease ,genetic testing ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Cancer Genetics ,Medicine ,Humans ,genetics ,Age of Onset ,Prospective cohort study ,skin and connective tissue diseases ,CHEK2 ,Genetics (clinical) ,Genetic testing ,medicine.diagnostic_test ,business.industry ,DNA, Neoplasm ,Sequence Analysis, DNA ,Ductal carcinoma ,medicine.disease ,Genes, p53 ,Checkpoint Kinase 2 ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cohort ,Mutation ,Female ,business - Abstract
BackgroundWhile the likelihood of identifying constitutional breast cancer-associated BRCA1, BRCA2 and TP53 pathogenic variants (PVs) increases with earlier diagnosis age, little is known about the correlation with age at diagnosis in other predisposition genes. Here, we assessed the contribution of known breast cancer-associated genes to very early onset disease.MethodsSequencing of BRCA1, BRCA2, TP53 and CHEK2 c.1100delC was undertaken in women with breast cancer diagnosed ≤30 years. Those testing negative were screened for PVs in a minimum of eight additional breast cancer-associated genes. Rates of PVs were compared with cases ≤30 years from the Prospective study of Outcomes in Sporadic vs Hereditary breast cancer (POSH) study.ResultsTesting 379 women with breast cancer aged ≤30 years identified 75 PVs (19.7%) in BRCA1, 35 (9.2%) in BRCA2, 22 (5.8%) in TP53 and 2 (0.5%) CHEK2 c.1100delC. Extended screening of 184 PV negative women only identified eight additional actionable PVs. BRCA1/2 PVs were more common in women aged 26–30 years than in younger women (p=0.0083) although the younger age group had rates more similar to those in the POSH cohort. Out of 26 women with ductal carcinoma in situ (DCIS) alone, most were high-grade and 11/26 (42.3%) had a PV (TP53=6, BRCA2=2, BRCA1=2, PALB2=1). This PV yield is similar to the 61 (48.8%) BRCA1/2 PVs identified in 125 women with triple-negative breast cancer. The POSH cohort specifically excluded pure DCIS which may explain lower TP53 PV rates in this group (1.7%).ConclusionThe rates of BRCA1, BRCA2 and TP53 PVs are high in very early onset breast cancer, with limited benefit from testing of additional breast cancer-associated genes.
- Published
- 2021
6. The spatial phenotype of genotypically distinct meningiomas demonstrate potential implications of the embryology of the meninges
- Author
-
D. G. R. Evans, Miriam J. Smith, Nicoletta Bobola, Claire O'Leary, Andrew T. King, Daniel M Fountain, Federico Roncaroli, and Omar N. Pathmanaban
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Brain tumor ,Context (language use) ,Review Article ,Biology ,Meningioma ,03 medical and health sciences ,0302 clinical medicine ,Chromosome instability ,otorhinolaryngologic diseases ,Genetics ,medicine ,neoplasms ,Cancer genetics ,Molecular Biology ,Meninges ,medicine.disease ,Hedgehog signaling pathway ,nervous system diseases ,030104 developmental biology ,medicine.anatomical_structure ,Differentiation ,030220 oncology & carcinogenesis ,Embryology ,Chromosome 22 - Abstract
Meningiomas are the most common primary brain tumor and their incidence and prevalence is increasing. This review summarizes current evidence regarding the embryogenesis of the human meninges in the context of meningioma pathogenesis and anatomical distribution. Though not mutually exclusive, chromosomal instability and pathogenic variants affecting the long arm of chromosome 22 (22q) result in meningiomas in neural-crest cell-derived meninges, while variants affecting Hedgehog signaling, PI3K signaling,TRAF7,KLF4, andPOLR2Aresult in meningiomas in the mesodermal-derived meninges of the midline and paramedian anterior, central, and ventral posterior skull base. Current evidence regarding the common pathways for genetic pathogenesis and the anatomical distribution of meningiomas is presented alongside existing understanding of the embryological origins for the meninges prior to proposing next steps for this work.
- Published
- 2020
7. The importance of genetic counseling and screening for people with pathogenic <scp> SMARCE1 </scp> variants: A family study
- Author
-
Alan Hewitt, Andrew T. King, Alireza Shoakazemi, D. Gareth Evans, Miriam J. Smith, Charlotte Hammerbeck-Ward, Scott A. Rutherford, Stavros Stivaros, Owen M. Thomas, Kenan Deniz, and Daniel du Plessis
- Subjects
0301 basic medicine ,business.industry ,Genetic counseling ,Context (language use) ,clear cell meningioma ,030105 genetics & heredity ,medicine.disease ,Bioinformatics ,SMARCE1 ,Asymptomatic ,Germline ,Meningioma ,03 medical and health sciences ,030104 developmental biology ,Genetics ,medicine ,Clear Cell Meningioma ,spinal tumor ,medicine.symptom ,Young adult ,business ,Genetics (clinical) ,Loss function - Abstract
Clear cell meningioma (CCM) is a rare variant of meningioma. In recent years, an association between cranial and spinal CCMs and germline loss of function mutations in the SMARCE1 gene (SWI/SNF chromatin remodeling complex subunit gene) has been discovered. We report a family with an incidental large spinal clear cell meningioma in a young adult following reflex screening for a germline loss of function pathogenic variant (PV) in the SMARCE1 gene. The index patient's mother and maternal grandfather were both also tested positive presymptomatically for SMARCE1. His mother developed intracranial and spinal meningiomas and his maternal grandfather developed a spinal CCM 4 years following a clear spinal MRI scan which required surgical excision. In this report we particularly emphasize the importance of genetic counseling and screening in siblings, parents and offspring of patients who are diagnosed with intracranial or spinal CCM in the context of SMARCE1 PVs. We recommend brain and spine Imaging screening of asymptomatic SMARCE1 PV carriers at least every 3 years, even if the baseline scan did not show any tumors.
- Published
- 2020
8. Incidence of mosaicism in 1055 de novo NF2 cases: much higher than previous estimates with high utility of next-generation sequencing
- Author
-
Omar N. Pathmanaban, Miriam J. Smith, Simon R. Freeman, Raji Anup, Mary Perry, D. Gareth Evans, Elaine F. Harkness, Emma Stapleton, Roger Laitt, Simon Tobi, Allyson Parry, Rupert Obholzer, Andrew T. King, Naomi L. Bowers, Philip T Smith, Shazia K. Afridi, Mark Kellett, Owen M. Thomas, Chris Duff, Grace Vassallo, Juliette Gair, Andrew J Wallace, Simon K W Lloyd, Scott A. Rutherford, Claire Hartley, Charlotte Hammerbeck-Ward, Stavros Stivaros, Patrick R. Axon, and Dorothy Halliday
- Subjects
0301 basic medicine ,Genetics ,Offspring ,Incidence (epidemiology) ,Heterozygote advantage ,Disease ,030105 genetics & heredity ,Biology ,medicine.disease ,DNA sequencing ,Neurofibromatosis type 2 ,03 medical and health sciences ,mosaicism ,030104 developmental biology ,NF2 ,otorhinolaryngologic diseases ,medicine ,LZTR1 ,Neurofibromatosis ,schwannoma ,Allele frequency ,Genetics (clinical) - Abstract
PURPOSE: To evaluate the incidence of mosaicism in de novo neurofibromatosis 2 (NF2).METHODS: Patients fulfilling NF2 criteria, but with no known affected family member from a previous generation (n = 1055), were tested for NF2 variants in lymphocyte DNA and where available tumor DNA. The proportion of individuals with a proven or presumed mosaic NF2 variant was assessed and allele frequencies of identified variants evaluated using next-generation sequencing.RESULTS: The rate of proven/presumed mosaicism was 232/1055 (22.0%). However, nonmosaic heterozygous pathogenic variants were only identified in 387/1055 (36.7%). When variant detection rates in second generation nonmosaics were applied to de novo cases, we assessed the overall probable mosaicism rate to be 59.7%. This rate differed by age from 21.7% in those presenting with bilateral vestibular schwannoma CONCLUSION: This study has identified a very high probable mosaicism rate in de novo NF2, probably making NF2 the condition with the highest expressed rate of mosaicism in de novo dominant disease that is nonlethal in heterozygote form. Risks to offspring are small and probably correlate with variant allele frequency detected in blood.
- Published
- 2020
9. Pathogenic noncoding variants in the neurofibromatosis and schwannomatosis predisposition genes
- Author
-
Cristina Perez-Becerril, D. Gareth Evans, and Miriam J. Smith
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Neurofibromatosis 1 ,Skin Neoplasms ,Neurofibromatoses ,intronic ,Biology ,Germline ,Pathogenesis ,Intergenic region ,Genetics ,medicine ,otorhinolaryngologic diseases ,Missense mutation ,Humans ,SMARCB1 ,Neurofibromatosis ,Schwannomatosis ,Gene ,pathogenic variants ,Genetics (clinical) ,schwannomatosis ,neurofibromatosis ,RNA-Binding Proteins ,noncoding ,SMARCB1 Protein ,medicine.disease ,MicroRNAs ,regulatory ,Neurilemmoma ,Transcription Factors - Abstract
Neurofibromatosis type 1 (NF1), type 2 (NF2), and schwannomatosis are a group of autosomal dominant disorders that predispose to the development of nerve sheath tumors. Pathogenic variants (PVs) that cause NF1 and NF2 are located in the NF1 and NF2 loci, respectively. To date, most variants associated with schwannomatosis have been identified in the SMARCB1 and LZTR1 genes, and a missense variant in the DGCR8 gene was recently reported to predispose to schwannomas. In spite of the high detection rate for PVs in NF1 and NF2 (over 90% of non-mosaic germline variants can be identified by routine genetic screening) underlying PVs for a proportion of clinical cases remain undetected. A higher proportion of non-NF2 schwannomatosis cases have no detected PV, with PVs currently only identified in around 70%–86% of familial cases and 30%–40% of non-NF2 sporadic schwannomatosis cases. A number of variants of uncertain significance have been observed for each disorder, many of them located in noncoding, regulatory, or intergenic regions. Here we summarize noncoding variants in this group of genes and discuss their established or potential role in the pathogenesis of NF1, NF2, and schwannomatosis.
- Published
- 2021
10. A mechanistic mathematical model of initiation and malignant transformation in sporadic vestibular schwannoma
- Author
-
X. Hoad, Ivana Bozic, Miriam J. Smith, D. G. R. Evans, and Chay Paterson
- Subjects
Vestibular system ,Cancer Research ,Mutation rate ,medicine.medical_treatment ,Point mutation ,Loss of Heterozygosity ,Neuroma, Acoustic ,Models, Theoretical ,Biology ,Schwannoma ,Malignancy ,medicine.disease ,Malignant transformation ,Radiation therapy ,Loss of heterozygosity ,Cell Transformation, Neoplastic ,Oncology ,medicine ,Cancer research ,Humans ,Neurilemmoma - Abstract
Background A vestibular schwannoma (VS) is a relatively rare, benign tumour of the eighth cranial nerve, often involving alterations to the gene NF2. Previous mathematical models of schwannoma incidence have not attempted to account for alterations in specific genes, and could not distinguish between nonsense mutations and loss of heterozygosity (LOH). Methods Here, we present a mechanistic approach to modelling initiation and malignant transformation in schwannoma. Each parameter is associated with a specific gene or mechanism operative in Schwann cells, and can be determined by combining incidence data with empirical frequencies of pathogenic variants and LOH. Results This results in new estimates for the base-pair mutation rate u = 4.48 × 10−10 and the rate of LOH = 2.03 × 10−6/yr in Schwann cells. In addition to new parameter estimates, we extend the approach to estimate the risk of both spontaneous and radiation-induced malignant transformation. Discussion We conclude that radiotherapy is likely to have a negligible excess risk of malignancy for sporadic VS, with a possible exception of rapidly growing tumours.
- Published
- 2021
11. Revised diagnostic criteria for neurofibromatosis type 1 and Legius syndrome: an international consensus recommendation
- Author
-
Matthias A. Karajannis, A Taylor, Diana Baralle, Rosalie E. Ferner, A Gomes, Dave Viskochil, J Toelen, Rianne Oostenbrink, Christopher L. Moertel, Laura Papi, Conxi Lázaro, H Wu, Michael D. Wilson, Shay Ben-Shachar, Pierre Wolkenstein, Sirkku Peltonen, Plotkin, P Joly, Dominique C. Pichard, Michael Fisher, Steinke-Lange, T Frébourg, P Ciavarelli, H Hanson, Mia MacCollin, I Blanco, D Bessis, Meena Upadhyaya, C Cassiman, Dusica Babovic-Vuksanovic, Riccardi, Juha Peltonen, James H. Tonsgard, B Poppe, Katharina Wimmer, M Larralde, P Pancza, A Heiberg, Bruce R. Korf, Mautner, D. G. R. Evans, Robert Listernick, Tena Rosser, S Barbarot, Eva Trevisson, D Stevenson, M Anten, Eduard Serra, Miriam J. Smith, Christopher J Hammond, Susan M Huson, Yemima Berman, Marco Giovannini, C Mallucci, Anat Stemmer-Rachamimov, G Tadini, Robert A. Avery, N Rezende, Nicole J. Ullrich, CO Hanemann, SM Stivaros, Hildegard Kehrer-Sawatzki, A Parry, D Kroshinsky, Maurizio Clementi, JT Jordan, A Varan, Joanne Ngeow, A Mueller, G Zadeh, Michel Kalamarides, D Halliday, M Link, Elizabeth K. Schorry, Roger J. Packer, Vanessa L. Merker, David H. Gutmann, Arthur S. Aylsworth, Karin Soares Gonçalves Cunha, V-F Mautner, Amanda L. Bergner, David A. Stevenson, Eric Legius, L Le, M Ruggieri, Fred G. Barker, Ludwine Messiaen, Jan M. Friedman, J. Blakeley, Kaleb Yohay, Katherine A. Rauen, LO Rodrigues, and Pediatrics
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Consensus ,Neurofibromatosis 1 ,Delphi method ,MEDLINE ,MUTATION ANALYSIS ,MOSAICISM ,Patient advocacy ,Article ,03 medical and health sciences ,0302 clinical medicine ,REVEALS ,SEQUENCE VARIANTS ,medicine ,Humans ,Cafe-au-Lait Spots ,Genetic Testing ,Medical physics ,Neurofibromatosis ,Genetics (clinical) ,Genetic testing ,Genetics & Heredity ,Legius syndrome ,Science & Technology ,IDENTIFICATION ,medicine.diagnostic_test ,business.industry ,medicine.disease ,GENE ,030104 developmental biology ,CHOROIDAL ABNORMALITIES ,030220 oncology & carcinogenesis ,business ,Life Sciences & Biomedicine - Abstract
PURPOSE: By incorporating major developments in genetics, ophthalmology, dermatology, and neuroimaging, to revise the diagnostic criteria for neurofibromatosis type 1 (NF1) and to establish diagnostic criteria for Legius syndrome (LGSS). METHODS: We used a multistep process, beginning with a Delphi method involving global experts and subsequently involving non-NF experts, patients, and foundations/patient advocacy groups. RESULTS: We reached consensus on the minimal clinical and genetic criteria for diagnosing and differentiating NF1 and LGSS, which have phenotypic overlap in young patients with pigmentary findings. Criteria for the mosaic forms of these conditions are also recommended. CONCLUSION: The revised criteria for NF1 incorporate new clinical features and genetic testing, whereas the criteria for LGSS were created to differentiate the two conditions. It is likely that continued refinement of these new criteria will be necessary as investigators (1) study the diagnostic properties of the revised criteria, (2) reconsider criteria not included in this process, and (3) identify new clinical and other features of these conditions. For this reason, we propose an initiative to update periodically the diagnostic criteria for NF1 and LGSS. ispartof: GENETICS IN MEDICINE vol:23 issue:8 pages:1506-1513 ispartof: location:United States status: published
- Published
- 2021
12. Gene Panel Testing for Breast Cancer Reveals Differential Effect of Prior BRCA1/2 Probability
- Author
-
Miriam J. Smith, Anthony Howell, D. Gareth Evans, William G. Newman, Elke M van Veen, Fiona Lalloo, Naomi L. Bowers, Elaine F. Harkness, Emma R. Woodward, Jamie M Ellingford, Sacha J Howell, and Andrew J Wallace
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,endocrine system diseases ,PALB2 ,CHEK2 ,Article ,panel test ,Breast cancer ,breast cancer ,Internal medicine ,Gene panel ,medicine ,PTEN ,skin and connective tissue diseases ,RC254-282 ,biology ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,ATM ,medicine.disease ,BRCA1 ,BRCA2 ,biology.protein ,Ovarian cancer ,business - Abstract
Whilst panel testing of an extended group of genes including BRCA1/2 is commonplace, these studies have not been subdivided by histiotype or by a priori BRCA1/2 probability. Patients with a breast cancer diagnosis undergoing extended panel testing were assessed for frequency of actionable variants in breast cancer genes other than BRCA1/2 by histiotype and Manchester score (MS) to reflect a priori BRCA1/2 likelihood. Rates were adjusted by prior testing for BRCA1/2 in an extended series. 95/1398 (6.3%) who underwent panel testing were found to be positive for actionable non-BRCA1/2 breast/ovarian cancer genes (ATM, BARD1, CDH1, CHEK2, PALB2, PTEN, RAD51C, RAD51D, TP53). As expected, PALB2, CHEK2 and ATM were predominant with 80-(5.3%). The highest rate occurred in Grade-3 ER+/HER2− breast cancers-(9.6%). Rates of non-BRCA actionable genes was fairly constant over all likelihoods of BRCA1/2 but adjusted rates were three times higher with MS <, 9 (BRCA1/2 = 1.5%, other = 4.7%), but was only 1.6% compared to 79.3% with MS ≥ 40. Although rates of detection of non-BRCA actionable genes are relatively constant across BRCA1/2 likelihoods this disguises an overall adjusted low frequency in high-likelihood families which have been heavily pre-tested for BRCA1/2. Any loss of detection sensitivity for BRCA1/2 actionable variants in breast cancer panels should lead to bespoke BRCA1/2 testing being conducted first.
- Published
- 2021
13. Identifying the deficiencies of current diagnostic criteria for neurofibromatosis 2 using databases of 2777 individuals with molecular testing
- Author
-
Omar N. Pathmanaban, Mary Perry, Patrick R. Axon, Dorothy Halliday, Miriam J. Smith, Owen M. Thomas, Roger Laitt, Allyson Parry, Juliette Gair, Mark Kellett, Elaine F. Harkness, Emma Stapleton, Rosalie E. Ferner, Simon R. Freeman, Andrew J Wallace, Simon K.L. Lloyd, Scott A. Rutherford, Naomi L. Bowers, Andrew T. King, Shazia K. Afridi, Raji Anup, Simon Tobi, D. Gareth Evans, and Charlotte Hammerbeck-Ward
- Subjects
Adult ,Male ,0301 basic medicine ,Ependymoma ,Neurofibromatosis 2 ,Schwannoma ,Adolescent ,Databases, Factual ,computer.software_genre ,Diagnosis, Differential ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Terminology as Topic ,otorhinolaryngologic diseases ,Humans ,Medicine ,Neurofibroma ,Neurofibromatosis type 2 ,Neurofibromatosis ,Sibling ,Child ,Schwannomatosis ,Genetics (clinical) ,Database ,business.industry ,Bilateral vestibular Schwannoma ,medicine.disease ,030104 developmental biology ,Molecular Diagnostic Techniques ,NF2 ,diagnostic criteria ,Female ,LZTR1 ,business ,computer ,030217 neurology & neurosurgery - Abstract
Purpose: We have evaluated deficiencies in existing diagnostic criteria for neurofibromatosis 2 (NF2). Methods: Two large databases of individuals fulfilling NF2 criteria (n=1361) and those tested for NF2 variants with criteria short of diagnosis (n=1416) were interrogated. We assessed the proportions meeting each diagnostic criterion with constitutional or mosaic NF2 variants and the Positive Predictive Value (PPV) with regard to definite diagnosis.Results: There was no evidence for usefulness of old criteria ‘glioma’ or ‘neurofibroma’. ‘Ependymoma’ had 100% PPV and high levels of confirmed NF2 diagnosis (67.7%). Those with bilateral vestibular schwannoma (VS) alone aged ≥60 years had the lowest confirmation rate (6.6%) and reduced PPV(80%). Siblings as a first-degree-relative, without an affected parent, had 0% PPV. All three individuals with unilateral VS and an affected sibling were proven not to have NF2. The biggest overlap was with LZTR1-associated schwannomatosis. In this category, seven individuals with unilateral VS plus ≥2 non-dermal schwannomas reduced PPV to 67%.Conclusion: The present study has confirmed important deficiencies in NF2 diagnostic criteria. The term ‘glioma’ should be dropped and replaced by ‘ependymoma’. Similarly ‘neurofibroma’ should be removed. Dropping ‘sibling’ from first-degree-relatives should be considered and testing of LZTR1 should be recommended for unilateral VS.
- Published
- 2019
14. Multiple primary malignancies associated with a germline SMARCB1 pathogenic variant
- Author
-
Patrick Shenjere, Richard W. Whitehouse, Naomi L. Bowers, James P Wylie, Judith Eelloo, Noel W. Clarke, Miriam J. Smith, D. Gareth Evans, Anthony J. Freemont, Chris Duff, Calvin Soh, Paul Hulse, John Ealing, and Mark Jones
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Horner Syndrome ,Skin Neoplasms ,Neurofibromatoses ,SMARCB1 ,030105 genetics & heredity ,Schwannoma ,Follicular lymphoma ,Neoplasms, Multiple Primary ,Lesion ,03 medical and health sciences ,0302 clinical medicine ,MPNST ,Biopsy ,Genetics ,Atypia ,Humans ,Medicine ,Neurofibroma ,Retroperitoneal Neoplasms ,Neurofibromatosis ,Schwannomatosis ,Genetics (clinical) ,medicine.diagnostic_test ,business.industry ,Malignancy ,SMARCB1 Protein ,Middle Aged ,medicine.disease ,Abdominal mass ,Neurofibromatosis type 2 ,Oncology ,Neurofibrosarcoma ,030220 oncology & carcinogenesis ,Female ,Radiology ,medicine.symptom ,Hemangioma ,business ,Neurilemmoma ,Neurofibromatosis type 1 - Abstract
A 51-year old presented with a 6-month history of increasing pelvic/lower back pain with nocturnal waking and episodes of anorexia and vomiting. Examination revealed right torticollis and Horner’s syndrome, and a large abdominal mass arising from the pelvis. Magnetic resonance and positron emission tomography imaging revealed A) a 14cm heterogeneous enhancing mass, abutting the left kidney with standardised uptake value max=2.9, B) a large heterogeneous enhancing pelvic mass C) mesenteric adenopathy standardised uptake value max=10.3 and D) 6cm right lung apex mass standardised uptake value max=4.3.Computerised tomography-guided biopsy of lesion A was reported as neurofibroma with occasional atypia, lesion B a benign uterine leiomyoma and lesion C follicular Lymphoma world health organisation Grade 2.Although she had been given the diagnosis of Neurofibromatosis Type-1 (NF1) 25-years previously following removal of an intradural extramedullary schwannoma she had no cutaneous stigmata of NF1. Genetic analysis of blood lymphocyte DNA identified a pathogenic variant in SMARCB1 confirming a diagnosis of schwannomatosis..Following 6-months chemotherapy for lymphoma, surgery was performed to remove lesion A. Histology revealed a malignant peripheral nerve sheath tumour with areas of low and high-grade change. An incidental, well-differentiated small bowel neuroendocrine carcinoma was also excised. Close surveillance continues with no recurrence after six years. This case study describes a novel finding of three separate synchronous primary malignancies in a patient with schwannomatosis and a proven SMARCB1 pathogenic variant.
- Published
- 2019
15. Typical 22q11.2 deletion syndrome appears to confer a reduced risk of schwannoma
- Author
-
Ludwine Messiaen, Barbara Rivera, Donna M. McDonald-McGinn, Alexandra Murray, Miriam J. Smith, William D. Foulkes, Rachel E A Irving, David A. Stevenson, D. Gareth Evans, and Cristina Perez-Becerril
- Subjects
Reduced risk ,Neurofibromatoses ,Population ,Schwannoma ,Bioinformatics ,Brief Communication ,Marfan Syndrome ,03 medical and health sciences ,0302 clinical medicine ,medicine ,DiGeorge Syndrome ,otorhinolaryngologic diseases ,Genetics ,Humans ,Deletion syndrome ,Schwannomatosis ,education ,Genetics (clinical) ,030304 developmental biology ,Genetic testing ,Tumors ,0303 health sciences ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,Neuroma, Acoustic ,medicine.disease ,Phenotype ,business ,030217 neurology & neurosurgery ,Neurilemmoma ,Genètica ,Transcription Factors - Abstract
Purpose The LZTR1 gene has been associated with schwannomatosis tumor predisposition and is located in a region that is deleted in the great majority (89%) of patients with 22q11.2 deletion syndrome (22q11.2DS). Since it is known that approximately 1 in 500 people in the general population will develop a sporadic schwannoma and there are no reports of the occurrence of schwannoma in 22q11.2DS, we investigated whether whole-gene deletion of LZTR1 occurs in schwannomatosis and assessed the risk of schwannoma in 22q11.2DS. Methods We assessed the genetic testing results for LZTR1-associated schwannomatosis and the clinical phenotypes of patients with 22q11.2DS. Results There were no reports of schwannoma in over 1,500 patients with 22q11.2DS. In addition, no patients meeting clinical diagnostic criteria for schwannomatosis had a whole-gene deletion in LZTR1. Only 1 patient in 110 with an apparently sporadic vestibular schwannoma had a constitutional whole-gene deletion of LZTR1. Conclusion People with a large 22q11.2 deletion may have a reduced risk of developing a schwannoma compared to the general population.
- Published
- 2021
16. Extended gene panel testing in lobular breast cancer
- Author
-
D. Gareth Evans, Miriam J. Smith, Elke M van Veen, William G. Newman, Emma R. Woodward, Anthony Howell, Jamie M Ellingford, Naomi L. Bowers, Andrew J Wallace, Elaine F. Harkness, Helen Byers, Sacha J Howell, and Fiona Lalloo
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,PALB2 ,Genes, BRCA2 ,Breast Neoplasms ,CDH1 ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Epidemiology ,Genetics ,medicine ,PTEN ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,CHEK2 ,Genetics (clinical) ,Germ-Line Mutation ,Genetic testing ,medicine.diagnostic_test ,biology ,business.industry ,Odds ratio ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Female ,business - Abstract
Purpose: Lobular breast cancer (LBC) accounts for ~ 15% of breast cancer. Here, we studied the frequency of pathogenic germline variants (PGVs) in an extended panel of genes in women affected with LBC. Methods: 302 women with LBC and 1567 without breast cancer were tested for BRCA1/2 PGVs. A subset of 134 LBC affected women who tested negative for BRCA1/2 PGVs underwent extended screening, including: ATM, CDH1, CHEK2, NBN, PALB2, PTEN, RAD50, RAD51D, and TP53.Results: 35 PGVs were identified in the group with LBC, of which 22 were in BRCA1/2. Ten actionable PGVs were identified in additional genes (ATM(4), CDH1(1), CHEK2(1), PALB2(2) and TP53(2)). Overall, PGVs in three genes conferred a significant increased risk for LBC. Odds ratios (ORs) were: BRCA1: OR = 13.17 (95%CI 2.83–66.38; P = 0.0017), BRCA2: OR = 10.33 (95%CI 4.58–23.95; P ATM: OR = 8.01 (95%CI 2.52–29.92; P = 0.0053). We did not detect an increased risk of LBC for PALB2, CDH1 or CHEK2. Conclusion: The overall PGV detection rate was 11.59%, with similar rates of BRCA1/2 (7.28%) PGVs as for other actionable PGVs (7.46%), indicating a benefit for extended panel genetic testing in LBC. We also report a previously unrecognised association of pathogenic variants in ATM with LBC.
- Published
- 2021
17. Disease course of neurofibromatosis type 2: a 30-year follow-up study of 353 patients seen at a single institution
- Author
-
Andrew T. King, Elaine F. Harkness, Emma M. Stapleton, D. Gareth Evans, Scott A. Rutherford, Grace Vassallo, Stavros Stivaros, Roger Laitt, Omar N. Pathmanaban, Claire Forde, Simon Kerrigan, Simon R. Freeman, Martin G. McCabe, Miriam J. Smith, Charlotte Hammerbeck-Ward, Simon K W Lloyd, John Paul Kilday, Catherine McBain, and Owen M. Thomas
- Subjects
Ependymoma ,Neurofibromatosis 2 ,Cancer Research ,Pediatrics ,medicine.medical_specialty ,Clinical Investigations ,Acoustic neuroma ,Asymptomatic ,Meningioma ,otorhinolaryngologic diseases ,Meningeal Neoplasms ,medicine ,Humans ,Neurofibromatosis type 2 ,Univariate analysis ,Proportional hazards model ,business.industry ,Neuroma, Acoustic ,medicine.disease ,Oncology ,Neurology (clinical) ,Age of onset ,medicine.symptom ,business ,Follow-Up Studies - Abstract
Background Limited data exist on the disease course of neurofibromatosis type 2 (NF2) to guide clinical trial design. Methods A prospective database of patients meeting NF2 diagnostic criteria, reviewed between 1990 and 2020, was evaluated. Follow-up to first vestibular schwannoma (VS) intervention and death was assessed by univariate analysis and stratified by age at onset, era referred, and inheritance type. Interventions for NF2-related tumors were assessed. Cox regression was performed to determine the relationship between individual factors from time of diagnosis to NF2-related death. Results Three hundred and fifty-three patients were evaluated. During 4643.1 follow-up years from diagnosis to censoring, 60 patients (17.0%) died. The annual mean number of patients undergoing VS surgery or radiotherapy declined, from 4.66 and 1.65, respectively, per 100 NF2 patients in 1990-1999 to 2.11 and 1.01 in 2010-2020, as the number receiving bevacizumab increased (2.51 per 100 NF2 patients in 2010-2020). Five patients stopped bevacizumab to remove growing meningioma or spinal schwannoma. 153/353 (43.3%) had at least one neurosurgical intervention/radiation treatment within 5 years of diagnosis. Patients asymptomatic at diagnosis had longer time to intervention and better survival compared to those presenting with symptoms. Those symptomatically presenting 40 years had poorer overall survival than those presenting at 26-39 years (P = .03 and P = .02, respectively) but those presenting between 16 and 39 had shorter time to VS intervention. Individuals with de novo constitutional variants had worse survival than those with de novo mosaic or inherited disease (P = .004). Conclusion Understanding disease course improves prognostication, allowing for better-informed decisions about care.
- Published
- 2020
18. Association between genetic polymorphisms and endometrial cancer risk:a systematic review
- Author
-
Neil A J Ryan, Anie Naqvi, D. Gareth Evans, Miriam J. Smith, Artitaya Lophatananon, Deborah J. Thompson, Cemsel Bafligil, Emma J Crosbie, Bafligil, Cemsel [0000-0002-2365-1194], Smith, Miriam J [0000-0002-3184-0817], Evans, D Gareth [0000-0002-8482-5784], Crosbie, Emma J [0000-0003-0284-8630], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,genetic epidemiology ,MEDLINE ,Kruppel-Like Transcription Factors ,Single-nucleotide polymorphism ,CINAHL ,Polymorphism, Single Nucleotide ,SOXC Transcription Factors ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,eIF-2 Kinase ,risk prediction ,0302 clinical medicine ,Aromatase ,single nucleotide polymorphism (SNP) ,systematic review ,Risk Factors ,Internal medicine ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Prospective Studies ,Genetics (clinical) ,Genetic association ,Hepatocyte Nuclear Factor 1-beta ,business.industry ,Endometrial cancer ,Proto-Oncogene Proteins c-mdm2 ,Publication bias ,medicine.disease ,HNF1B ,Endometrial Neoplasms ,030104 developmental biology ,Genetic epidemiology ,030220 oncology & carcinogenesis ,endometrial cancer ,Female ,business ,Genome-Wide Association Study - Abstract
Funder: national institute of health research, INTRODUCTION: Endometrial cancer is one of the most commonly diagnosed cancers in women. Although there is a hereditary component to endometrial cancer, most cases are thought to be sporadic and lifestyle related. The aim of this study was to systematically review prospective and retrospective case-control studies, meta-analyses and genome-wide association studies to identify genomic variants that may be associated with endometrial cancer risk. METHODS: We searched MEDLINE, Embase and CINAHL from 2007 to 2019 without restrictions. We followed PRISMA 2009 guidelines. The search yielded 3015 hits in total. Following duplicate exclusion, 2674 abstracts were screened and 453 full-texts evaluated based on our pre-defined screening criteria. 149 articles were eligible for inclusion. RESULTS: We found that single nucleotide polymorphisms (SNPs) in HNF1B, KLF, EIF2AK, CYP19A1, SOX4 and MYC were strongly associated with incident endometrial cancer. Nineteen variants were reported with genome-wide significance and a further five with suggestive significance. No convincing evidence was found for the widely studied MDM2 variant rs2279744. Publication bias and false discovery rates were noted throughout the literature. CONCLUSION: Endometrial cancer risk may be influenced by SNPs in genes involved in cell survival, oestrogen metabolism and transcriptional control. Larger cohorts are needed to identify more variants with genome-wide significance.
- Published
- 2020
19. Sporadic vestibular schwannoma: a molecular testing summary
- Author
-
Adam Shaw, Simon R. Freeman, Miriam J. Smith, Amy E Taylor, Andrew T. King, Philip T Smith, Omar N. Pathmanaban, Simon Tobi, Claire Hartley, Naomi L. Bowers, D. Gareth Evans, Dorothy Halliday, Andrew J Wallace, Simon K W Lloyd, Emma Stapleton, Scott A. Rutherford, Katherine V Sadler, and Charlotte Hammerbeck-Ward
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Neurofibromatosis 2 ,Neurology ,Skin Neoplasms ,Adolescent ,Neurofibromatoses ,Schwannoma ,Germline ,Diagnosis, Differential ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Internal medicine ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Neurofibromatosis type 2 ,SMARCB1 ,Schwannomatosis ,Child ,Genetics (clinical) ,Genetic testing ,Aged ,Neurofibromin 2 ,medicine.diagnostic_test ,business.industry ,Infant ,Neuroma, Acoustic ,SMARCB1 Protein ,Middle Aged ,medicine.disease ,030220 oncology & carcinogenesis ,Child, Preschool ,Female ,Neurosurgery ,business ,030217 neurology & neurosurgery ,Neurilemmoma ,Transcription Factors - Abstract
ObjectivesCases of sporadic vestibular schwannoma (sVS) have a low rate of association with germline pathogenic variants. However, some individuals with sVS can represent undetected cases of neurofibromatosis type 2 (NF2) or schwannomatosis. Earlier identification of patients with these syndromes can facilitate more accurate familial risk prediction and prognosis.MethodsCases of sVS were ascertained from a local register at the Manchester Centre for Genomic Medicine. Genetic analysis was conducted in NF2 on blood samples for all patients, and tumour DNA samples when available. LZTR1 and SMARCB1 screening was also performed in patient subgroups.ResultsAge at genetic testing for vestibular schwannoma (VS) presentation was younger in comparison with previous literature, a bias resulting from updated genetic testing recommendations. Mosaic or constitutional germline NF2 variants were confirmed in 2% of patients. Pathogenic germline variants in LZTR1 were found in 3% of all tested patients, with a higher rate of 5% in patients SMARCB1 variants were identified within the cohort. Considering all individuals who received tumour DNA analysis, 69% of patients were found to possess two somatic pathogenic NF2 variants, including those with germline LZTR1 pathogenic variants.ConclusionsUndiagnosed schwannoma predisposition may account for a significant minority of apparently sVS cases, especially at lower presentation ages. Loss of NF2 function is a common event in VS tumours and may represent a targetable common pathway in VS tumourigenesis. These data also support the multi-hit mechanism of LZTR1-associated VS tumourigenesis.
- Published
- 2020
20. Risk of Contralateral Breast Cancer in Women with and without Pathogenic Variants in BRCA1, BRCA2, and TP53 Genes in Women with Very Early-Onset (<36 Years) Breast Cancer
- Author
-
Marta Pereira, Sacha J Howell, Anthony Howell, D. Gareth Evans, Fiona Lalloo, Miriam J. Smith, Andrew J Wallace, Elaine F. Harkness, Emma R. Woodward, Zerin Hyder, Naomi L. Bowers, and William G. Newman
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,brca1 ,endocrine system diseases ,medicine.medical_treatment ,Population ,Early-onset breast cancer ,contralateral ,Contralateral ,lcsh:RC254-282 ,Article ,Contralateral breast cancer ,03 medical and health sciences ,Breast cancer ,0302 clinical medicine ,breast cancer ,brca2 ,Internal medicine ,medicine ,TP53 Genes ,TP53 ,Family history ,Risk factor ,education ,skin and connective tissue diseases ,pathogenic variants ,education.field_of_study ,business.industry ,Pathogenic variants ,tp53 ,medicine.disease ,BRCA1 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,BRCA2 ,030104 developmental biology ,early-onset breast cancer ,Oncology ,030220 oncology & carcinogenesis ,Life expectancy ,business ,Mastectomy - Abstract
Early age at diagnosis of breast cancer is a known risk factor for hereditary predisposition and some studies show a high risk of contralateral breast cancer in BRCA1 carriers diagnosed at very young ages. However, little is published on the risk of TP53 carriers. 397 women with breast cancer diagnosed BRCA1, BRCA2, and TP53 genes was carried out alongside tests for copy number for PV on all referred women. Rates of contralateral breast cancer were censored at death, last assessment, or risk-reducing mastectomy. In total, 47 TP53, 218 BRCA1, and 132 BRCA2 PV carriers were identified with breast cancer diagnosed aged 35 years and under, as well as a representative sample of 261 not known to carry a PV in BRCA1, BRCA2, and TP53. Annual rates of contralateral breast cancer (and percentage of synchronous breast cancers) were TP53: 7.03% (4.3%), BRCA1: 3.57% (1.8%), and BRCA2: 2.63% (1.5%). In non-PV carriers, contralateral rates in isolated presumed/tested non-carrier cases with no family history were 0.56%, and for those with a family history, 0.69%. Contralateral breast cancer rates are substantial in TP53, BRCA1, and BRCA2 PV carriers diagnosed with breast cancer aged 35 and under. Women need to be advised to help make informed decisions on contralateral mastectomy, guided by life expectancy from their index tumor.
- Published
- 2020
21. Familial unilateral vestibular schwannoma is rarely caused by inherited variants in the NF2 gene
- Author
-
Mark Kellett, Omar N. Pathmanaban, Andrew T. King, Charlotte Hammerbeck-Ward, Jemma Bischetsrieder, Patrick R. Axon, Scott A. Rutherford, Miriam J. Smith, Simon R. Freeman, Jaishri O. Blakeley, D. Gareth Evans, Owen M. Thomas, Claire Hartley, Roger Laitt, Andrew J Wallace, and Simon K W Lloyd
- Subjects
0301 basic medicine ,Adult ,Pediatrics ,medicine.medical_specialty ,Otology/Neurotology ,Adolescent ,Context (language use) ,Nf2 gene ,Retrospective database ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Older patients ,vestibular schwannoma ,Genes, Neurofibromatosis 2 ,Original Reports ,otorhinolaryngologic diseases ,Medicine ,Humans ,Neurofibromatosis type 2 ,Child ,Uncertain significance ,Aged ,Retrospective Studies ,business.industry ,Unilateral vestibular schwannoma ,familial ,Unilateral ,Neuroma, Acoustic ,Middle Aged ,medicine.disease ,Clinical Practice ,030104 developmental biology ,Otorhinolaryngology ,NF2 ,Child, Preschool ,LZTR1 ,business ,030217 neurology & neurosurgery - Abstract
Objectives/hypothesis Unilateral vestibular schwannoma (VS) occurs with a lifetime risk of around 1 in 1,000 and is due to inactivation of the NF2 gene, either somatically or from a constitutional mutation. It has been postulated that familial occurrence of unilateral VS occurs more frequently than by chance, but no causal mechanism has been confirmed. Study design Retrospective database analysis. Methods The likelihood of chance occurrence of unilateral VS, or occurring in the context of neurofibromatosis type 2 (NF2), was assessed using national UK audit data and data from the national NF2 database. Families with familial unilateral VS (occurrence in first- and second-degree relatives) were assessed for constitutional NF2 and LZTR1 genetic variants, and where possible the tumor was also analyzed. Results Approximately 1,000 cases of unilateral VS occurred annually in the United Kingdom between 2013 and 2016. Of these, 2.5 may be expected to have a first-degree relative who had previously developed a unilateral VS. The likelihood of this occurring in NF2 was considered to be as low as 0.05 annually. None of 28 families with familial unilateral VS had a constitutional NF2 intragenic variant, and in nine cases where the VS was analyzed, both mutational events in NF2 were identified and excluded from the germline. Only three variants of uncertain significance were found in LZTR1. Conclusions Familial occurrence of unilateral VS is very unlikely to be due to a constitutional NF2 or definitely pathogenic LZTR1 variant. The occurrence of unilateral VS in two or more first-degree relatives is likely due to chance. This phenomenon may well increase in clinical practice with increasing use of cranial magnetic resonance imaging in older patients. Level of evidence 2b Laryngoscope, 129:967-973, 2019.
- Published
- 2018
22. Schwannomatosis: a genetic and epidemiological study
- Author
-
Simon R. Freeman, Owen M. Thomas, Claire Hartley, Chris Duff, Roger Laitt, Omar N. Pathmanaban, Miriam J. Smith, Andrew J Wallace, D. Gareth Evans, Simon K W Lloyd, Rosalie E. Ferner, John Ealing, Naomi L. Bowers, Scott A. Rutherford, Amy Taylor, Charlotte Hammerbeck-Ward, Dorothy Halliday, Mark Kellett, Simon Tobi, Andrew T. King, and Elaine F. Harkness
- Subjects
Adult ,Male ,Neurofibromatosis 2 ,Pediatrics ,medicine.medical_specialty ,Skin Neoplasms ,Adolescent ,Databases, Factual ,Neurofibromatoses ,Population ,Prevalence ,SMARCB1 ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,vestibular schwannoma ,Epidemiology ,otorhinolaryngologic diseases ,medicine ,Humans ,education ,Schwannomatosis ,Aged ,Aged, 80 and over ,schwannomatosis ,Neurofibromin 2 ,education.field_of_study ,business.industry ,Incidence ,Incidence (epidemiology) ,Mean age ,SMARCB1 Protein ,Middle Aged ,medicine.disease ,Psychiatry and Mental health ,England ,030220 oncology & carcinogenesis ,Life expectancy ,Female ,Surgery ,Neurology (clinical) ,LZTR1 ,business ,Neurilemmoma ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
ObjectivesSchwannomatosis is a dominantly inherited condition predisposing to schwannomas of mainly spinal and peripheral nerves with some diagnostic overlap with neurofibromatosis-2 (NF2), but the underlying epidemiology is poorly understood. We present the birth incidence and prevalence allowing for overlap with NF2.MethodsSchwannomatosis and NF2 cases were ascertained from the Manchester region of England (population=4.8 million) and from across the UK. Point prevalence and birth incidence were calculated from regional birth statistics. Genetic analysis was also performed on NF2, LZTR1 and SMARCB1 on blood and tumour DNA samples when available.ResultsRegional prevalence for schwannomatosis and NF2 were 1 in 126 315 and 50 500, respectively, with calculated birth incidences of 1 in 68 956 and 1 in 27 956. Mosaic NF2 causes a substantial overlap with schwannomatosis resulting in the misdiagnosis of at least 9% of schwannomatosis cases. LZTR1-associated schwannomatosis also causes a small number of cases that are misdiagnosed with NF2 (1%–2%), due to the occurrence of a unilateral vestibular schwannoma. Patients with schwannomatosis had lower numbers of non-vestibular cranial schwannomas, but more peripheral and spinal nerve schwannomas with pain as a predominant presenting symptom. Life expectancy was significantly better in schwannomatosis (mean age at death 76.9) compared with NF2 (mean age at death 66.2; p=0.004).ConclusionsWithin the highly ascertained North-West England population, schwannomatosis has less than half the birth incidence and prevalence of NF2.
- Published
- 2018
23. Abstract PD1-05: Transgenerational epigenetic silencing of BRCA1 due to a germline variant unmasks a new mechanism for familial breast and ovarian cancer
- Author
-
Helen Byers, Andrew J Wallace, Miriam J. Smith, D G R Evans, F Lalloo, William G. Newman, and EM van Veen
- Subjects
Cancer Research ,Oncology ,Transgenerational epigenetics ,Mechanism (biology) ,Cancer research ,medicine ,Biology ,Ovarian cancer ,medicine.disease ,Germline ,Epigenetic silencing - Abstract
In families with multiple affected individuals with early-onset breast/ovarian cancer pathogenic variants in BRCA1 or BRCA2 are identified in approximately 20% of the cases. Extensive efforts have been made to identify additional highly penetrant breast cancer genes or alternative mutational mechanisms affecting BRCA1 and BRCA2 to explain the missing heritability. It has been proposed that part of the missing heritability may be explained by gene silencing due to promoter methylation of cancer associated genes, as described in colorectal cancer (MLH1 and MSH2). Here, for the first time, we report two independent families with multiple individuals affected by breast and ovarian cancer with transgenerational promoter methylation of BRCA1. RNA analysis of BRCA1 in the germline of breast/ovarian families, previously found to be BRCA1/2 mutation negative by Sanger sequencing and copy number analysis, identified two families with allelic loss of expression. To investigate the mechanism of transcriptional silencing, a total of 14 affected and unaffected family members from these two families were tested for BRCA1 promoter methylation by pyrosequencing in blood, buccal, and hair follicle cells. Allele specific methylation was determined by clonal bisulphite sequencing. BRCA1 promoter methylation in all three germ layers was present in 11 of 14 family members. Of the 7 women with promoter methylation five were affected with grade 3 breast/high grade serous ovarian cancer. The four males with BRCA1 promoter methylation had no history of cancer. Methylation levels were ˜50%, consistent with the silencing of one allele detected in RNA in these family members. Clonal bisulphite sequencing of an affected family member of each family confirmed that the alternative allele was specifically methylated. Interestingly, in both families the methylation pattern of the BRCA1 promoter segregated with the same novel heterozygous variant in the 5'UTR of BRCA1. These results indicate a novel mechanism for familial breast/ovarian cancer, caused by epigenetic silencing of one allele by transgenerational hypermethylation of the BRCA1 promoter, secondary to a variant in cis of BRCA1. We propose that methylation analyses are indicated in all families affected by early onset breast/ovarian cancer where standard mutation screening of BRCA1/2 has not identified a causative variant. Citation Format: van Veen EM, Smith MJ, Byers HJ, Wallace AJ, Lalloo FI, Newman WG, Evans DGR. Transgenerational epigenetic silencing of BRCA1 due to a germline variant unmasks a new mechanism for familial breast and ovarian cancer [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr PD1-05.
- Published
- 2018
24. A Novel PTCH1 Frameshift Mutation Leading to Nevoid Basal Cell Carcinoma Syndrome
- Author
-
Gareth Evans, Timur Tuncalı, Miriam J. Smith, Pelin Ertop, Ceren D. Durmaz, and Bengü Nisa Akay
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Macrocephaly ,Nevoid basal-cell carcinoma syndrome ,Biology ,medicine.disease ,Frameshift mutation ,Falx cerebri ,PTCH2 ,stomatognathic diseases ,03 medical and health sciences ,Frontal Bossing ,030104 developmental biology ,0302 clinical medicine ,PTCH1 ,030220 oncology & carcinogenesis ,Genetics ,medicine ,medicine.symptom ,Keratocyst ,Molecular Biology ,Genetics (clinical) - Abstract
Nevoid basal cell carcinoma syndrome (NBCCS), also known as Gorlin syndrome, is a rare multisystemic autosomal dominant disorder typically presenting with cutaneous basal cell carcinomas, multiple keratocysts, and skeletal anomalies. NBCCS is caused by heterozygous mutations in the PTCH1 gene in chromosome 9q22, in the PTCH2 gene in 1p34, or the SUFU gene in 10q24.32. Here, we report on an 18-month-old boy presenting with medulloblastoma, frontal bossing, and multiple skeletal anomalies and his father who has basal cell carcinomas, palmar pits, macrocephaly, bifid ribs, calcification of falx cerebri, and a history of surgery for odontogenic keratocyst. These clinical findings were compatible with the diagnosis of NBCCS, and a novel mutation, c.1249delC; p.Gln417Lysfs*15, was found in PTCH1 causing a premature stop codon.
- Published
- 2018
25. Characterization of age-dependent and progressive cortical neuronal degeneration in presenilin conditional mutant mice.
- Author
-
Mary Wines-Samuelson, Eva C Schulte, Miriam J Smith, Chiye Aoki, Xinran Liu, Raymond J Kelleher, and Jie Shen
- Subjects
Medicine ,Science - Abstract
Presenilins are the major causative genes of familial Alzheimer's disease (AD). Our previous study has demonstrated essential roles of presenilins in memory and neuronal survival. Here, we explore further how loss of presenilins results in age-related, progressive neurodegeneration in the adult cerebral cortex, where the pathogenesis of AD occurs. To circumvent the requirement of presenilins for embryonic development, we used presenilin conditional double knockout (Psen cDKO) mice, in which presenilin inactivation is restricted temporally and spatially to excitatory neurons of the postnatal forebrain beginning at 4 weeks of age. Increases in the number of degenerating (Fluoro-Jade B+, 7.6-fold) and apoptotic (TUNEL+, 7.4-fold) neurons, which represent approximately 0.1% of all cortical neurons, were first detected at 2 months of age when there is still no significant loss of cortical neurons and volume in Psen cDKO mice. By 4 months of age, significant loss of cortical neurons (approximately 9%) and gliosis was found in Psen cDKO mice. The apoptotic cell death is associated with caspase activation, as shown by increased numbers of cells immunoreactive for active caspases 9 and 3 in the Psen cDKO cortex. The vulnerability of cortical neurons to loss of presenilins is region-specific with cortical neurons in the lateral cortex most susceptible. Compared to the neocortex, the increase in apoptotic cell death and the extent of neurodegeneration are less dramatic in the Psen cDKO hippocampus, possibly in part due to increased neurogenesis in the aging dentate gyrus. Neurodegeneration is also accompanied with mitochondrial defects, as indicated by reduced mitochondrial density and altered mitochondrial size distribution in aging Psen cortical neurons. Together, our findings show that loss of presenilins in cortical neurons causes apoptotic cell death occurring in a very small percentage of neurons, which accumulates over time and leads to substantial loss of cortical neurons in the aging brain. The low occurrence and significant delay of apoptosis among cortical neurons lacking presenilins suggest that loss of presenilins may induce apoptotic neuronal death through disruption of cellular homeostasis rather than direct activation of apoptosis pathways.
- Published
- 2010
- Full Text
- View/download PDF
26. CTNI-54. A SINGLE ARM PHASE II STUDY OF THE DUAL MTORC1/MTORC2 INHIBITOR VISTUSERTIB PROVIDED FOR SPORADIC PATIENTS WITH GRADE II-III MENINGIOMAS THAT RECUR OR PROGRESS AFTER SURGERY AND RADIATION
- Author
-
Patrick Y. Wen, Fred G. Barker, Vijaya Ramesh, Anat Stemmer-Rachamimov, Alona Muzikansky, Miriam J. Smith, Justin T. Jordan, Roberta L. Beauchamp, Priya Kumthekar, Elizabeth R. Gerstner, and Scott R. Plotkin
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Surrogate endpoint ,Nausea ,Phases of clinical research ,medicine.disease ,Meningioma ,Oncology ,Troponin I ,Vomiting ,Medicine ,Neurology (clinical) ,Radiology ,Progression-free survival ,medicine.symptom ,business ,Adverse effect - Abstract
Grade II/III meningiomas have increased rates of recurrence with no approved medical therapies. The historical progression-free survival at 6 months (PFS-6) is 25% with rates >35% declared of interest for drug development. NF2 gene inactivation occurs in about half of meningiomas. Based on our studies showing mTORC1 and mTORC2/SGK1 pathway activation in NF2-deficient meningiomas and the paradoxical activation of the mTORC2/AKT pathway, we hypothesized that mTORC1/mTORC2 inhibitors would be active in meningiomas. We studied the effect of vistusertib in patients with progressive/recurrent grade II/III meningiomas (NCT03071874). Vistusertib was administered orally at 125mg twice daily on two consecutive days each week. MRIs were obtained every 56 days. Tumor size was defined as the largest cross-sectional area. Progression was defined as ≥ 25% increase in the sum of products of all measurable lesions over smallest sum observed. The primary endpoint was PFS-6. Secondary endpoints included toxicity, radiographic response, and correlative studies including immunohistochemistry for mTORC1/2 pathway activation and genetic biomarkers. Twenty-eight patients (13 female, median age 58 years, median KPS 80%) were enrolled. Median tumor size was 4.4cm; 71% were grade II and 50% harbored pathogenic NF2 variants. Four patients discontinued treatment voluntarily and 1 each withdrew for intercurrent illness and non-compliance. PFS-6 is 47% (CI, 26%-65%) and OS-12 is 72% (95%CI, 48%-86%). PFS but not OS was shorter for patients with grade 3 meningiomas; there was no difference in PFS/OS between genetic groups. Adverse events at least possibly related to vistusertib with frequency >10% include nausea, fatigue, hypophosphatemia, diarrhea, anorexia, dry mouth, hypertriglyceridemia, hypertension, vomiting, increased ALT, constipation, and weight loss. Vistusertib treatment was associated with a PFS-6 rate exceeding the target of 35% for recurrent high-grade meningioma. Adverse events were tolerable in this patient population. These data support the continued development of mTORC1/2 inhibitors in this setting.
- Published
- 2021
27. RARE-21. CANCER SPECTRUM IN GERMLINE SUFU MUTATION CARRIERS: A COLLABORATIVE PROJECT OF THE SIOPE HOST GENOME WORKING GROUP
- Author
-
Hélène Zattara, Stefan M. Pfister, Pascale Varlet, Laurence Brugières, Jacques Grill, Christian P. Kratz, Stéphanie Puget, Christelle Dufour, Franck Bourdeaut, David R. Jones, Marie Agnès Rame Collonge, Paul A. Northcott, Nicola Dikow, Léa Guerrini-Rousseau, Lucie Lafay-Cousin, Nicolas Sevenet, Till Milde, Andrey Korshunov, Miriam J. Smith, Eric Sariban, Giles W. Robinson, Dominik Sturm, Julien Masliah, Majoline Jongmans, Kristian W. Pajtler, Amar Gajjar, Sebastian M. Waszak, Olivier Delattre, Saskia Hopman, Gareth Evans, and Steffen Hirsch
- Subjects
Genetics ,Cancer Research ,Host genome ,Genetic inheritance ,Cancer ,Biology ,medicine.disease ,Genome ,Germline ,Oncology ,Mutation (genetic algorithm) ,medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Cancer risk ,Craniopharyngioma and Rare Tumors - Abstract
BACKGROUND Little is known about cancer risk associated with pathogenic germline SUFU variants. METHODS Data of all previously published and 25 still unpublished patients with a pathogenic germline SUFU mutation were compiled. RESULTS 124 patients in 67 families were identified, most of them ascertained after the occurrence of a medulloblastoma (MB) or as part of Gorlin syndrome cohorts. Overall, 30 patients were healthy carriers and 94 patients developed a total of 129 tumors (up to 4 tumors/patient): 68 MBs, always as first tumor (median age at diagnosis: 1.5yr [0.1–5]), 22 patients with at least 1 basal cell carcinoma (BCC) (median 10/patient) (median age at first BCC: 43yr, [17–52]), 15 meningiomas (median age 43yr, [13–72]), 7 ovarian stromal/fibrous tumors (median age 12yr [5–34]), and 17 other tumors including 5 sarcomas (median age: 50yr [7–79]). Median age at last follow-up was 30yr. Nineteen patients died, including 11 from MB. Second malignancies were diagnosed in 21 patients including 13 in MB survivors. Mutations were inherited in 58/66 (88%) of cases in which inheritance could be tested and de novo in 8. In 6/67 families (9%), >2 children were diagnosed with a MB. CONCLUSION In this large cohort of germline SUFU mutation carriers, MB in infants is the most frequent tumor but the spectrum also includes typical Gorlin syndrome tumors (BCC, meningiomas, and ovarian stromal/fibrous tumors) either as first tumors or as second malignancies. This broad tumor spectrum and the high risk of second malignancies justify the implementation of specific cancer surveillance programs.
- Published
- 2020
28. First evidence of genotype–phenotype correlations in Gorlin syndrome
- Author
-
Deemesh Oudit, D. Gareth Evans, David Rutkowski, William G. Newman, Ernest Allan, Miriam J. Smith, and John T. Lear
- Subjects
Adult ,Male ,0301 basic medicine ,endocrine system ,Pathology ,medicine.medical_specialty ,PTCH1 ,SUFU ,Adolescent ,Biology ,medicine.disease_cause ,Germline ,Meningioma ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Missense mutation ,Cerebellar Neoplasms ,Child ,Genetic Association Studies ,Germ-Line Mutation ,Genetics (clinical) ,Aged ,Aged, 80 and over ,Medulloblastoma ,Ovarian fibroma ,Mutation ,Infant ,Basal Cell Nevus Syndrome ,Middle Aged ,medicine.disease ,Phenotype ,Gorlin syndrome ,Patched-1 Receptor ,Repressor Proteins ,stomatognathic diseases ,030104 developmental biology ,Child, Preschool ,030220 oncology & carcinogenesis ,Female - Abstract
Background Gorlin syndrome (GS) is an autosomal dominant syndrome characterised by multiple basal cell carcinomas (BCCs) and an increased risk of jaw cysts and early childhood medulloblastoma. Heterozygous germline variants in PTCH1 and SUFU encoding components of the Sonic hedgehog pathway explain the majority of cases. Here, we aimed to delineate genotype–phenotype correlations in GS. Methods We assessed genetic and phenotypic data for 182 individuals meeting the diagnostic criteria for GS (median age: 47.1; IQR: 31.1–61.1). A total of 126 patients had a heterozygous pathogenic variant, 9 had SUFU pathogenic variants and 46 had no identified mutation. Results Patients with variants were more likely to be diagnosed earlier (p=0.02), have jaw cysts (p=0.002) and have bifid ribs (p=0.003) or any skeletal abnormality (p=0.003) than patients with no identified mutation. Patients with a missense variant in PTCH1 were diagnosed later (p=0.03) and were less likely to develop at least 10 BCCs and jaw cysts than those with other pathogenic PTCH1 variants (p=0.03). Patients with SUFU pathogenic variants were significantly more likely than those with PTCH1 pathogenic variants to develop a medulloblastoma (p=0.009), a meningioma (p=0.02) or an ovarian fibroma (p=0.015), but were less likely to develop a jaw cyst (p=0.0004). Conclusion We propose that the clinical heterogeneity of GS can in part be explained by the underlying or SUFU variant.
- Published
- 2017
29. SMARCE1mutation screening in classification of clear cell meningiomas
- Author
-
Miriam J. Smith, Jung-Il Lee, Soomin Ahn, Yeon-Lim Suh, Daniel du Plessis, and Michael Bulman
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Histology ,Adolescent ,Chromosomal Proteins, Non-Histone ,DNA Mutational Analysis ,Meningothelial Meningioma ,Biology ,medicine.disease_cause ,meningioma ,Pathology and Forensic Medicine ,Meningioma ,Young Adult ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Meningeal Neoplasms ,otorhinolaryngologic diseases ,Clear Cell Meningioma ,medicine ,Humans ,Child ,neoplasms ,Sanger sequencing ,Mutation ,General Medicine ,Middle Aged ,clear cell meningioma ,medicine.disease ,Immunohistochemistry ,SMARCE1 ,nervous system diseases ,DNA-Binding Proteins ,030220 oncology & carcinogenesis ,symbols ,Female ,Multiplex Polymerase Chain Reaction ,030217 neurology & neurosurgery ,Clear cell - Abstract
AIMS: Clear cell meningioma is a rare subtype of meningioma and shows not only unusual histology, but also unique clinical features. Recently, SMARCE1 mutations have been shown to cause spinal and cranial clear cell meningiomas. We present 12 cases which were diagnosed with a clear cell meningioma in a single institution between 1997 and 2014, and investigate their SMARCE1 mutation status.METHODS AND RESULTS: To investigate the SMARCE1 mutation status of these tumours, we used a combination of Sanger sequencing and multiplex ligation dependent probe amplification analysis and also performed SMARCE1 immunohistochemical staining. We found both SMARCE1 mutational hits, including novel SMARCE1 mutations, in six out of eight tested patients. Immunohistochemical analysis showed loss of SMARCE1 protein staining in all but two cases. Two individuals who were originally diagnosed with clear cell meningioma were negative for SMARCE1 mutations, but tested positive for NF2 mutations. As a result, these two tumours were reanalyzed and eventually reclassified, as a microcystic and a mixed growth pattern meningioma with focal clear cell areas, respectively.CONCLUSIONS: These results expand the spectrum of pathogenic variants in SMARCE1 and show that mutation screening can help facilitate meningioma classification. This may have implications for prognosis and future clinical management of patients, since clear cell meningiomas are classed as grade II tumours, while microcystic and meningothelial meningiomas are classed as grade I. This article is protected by copyright. All rights reserved.
- Published
- 2017
30. A novel c.885+1G>A splicing variant in exon 9 of the NF2 gene shows a delayed mild presentation with a predilection for spinal ependymomas
- Author
-
D G R Evans, Alejandro Feria, Alireza Shoakazemi, Scott A. Rutherford, Andrew T. King, Charlotte Hammerbeck-Ward, and Miriam J. Smith
- Subjects
Ependymoma ,congenital, hereditary, and neonatal diseases and abnormalities ,Pathology ,medicine.medical_specialty ,Splice site mutation ,business.industry ,Bilateral vestibular Schwannoma ,Autosomal dominant trait ,Schwannoma ,medicine.disease ,Meningioma ,Exon ,otorhinolaryngologic diseases ,medicine ,Neurofibromatosis type 2 ,business - Abstract
Neurofibromatosis Type 2 (NF2) is a rare autosomal dominant disease caused by pathological variants of the tumor suppressor gene NF2 classically manifesting as bilateral vestibular schwannoma, intracranial and spinal neoplasms such as meningioma and ependymoma, and posterior subcapsular and cortical wedge lens opacities, with an average onset of 18-24 years of age. Approximately 50% of patients inherit the disorder from an affected parent with the remainder resulting from de novo mutations or genetic mosaicism. The most common variants of NF2 are due to C>T transitions resulting in nonsense pathological variants. Individuals with splice-site variants of NF2 display varied phenotypes. Here we present a family with NF2 as the result of an apparently de novo c.885+1G>A splicing variant in exon 9 of the NF2 gene not previously described in the literature. This variant appears to be associated with an extremely mild phenotype with regards to vestibular schwannomas, other schwannoma disease and meningioma, instead exhibiting a delayed presentation with a predilection for ependymomas. The non-classical presentation of this NF2 splicing variant illustrates the importance of keeping NF2 in the differential diagnosis in patients with multiple spinal ependymomas with delayed onset and may warrant genetic testing for NF2 variants in such patients.
- Published
- 2019
31. Author response for 'Epithelial ovarian cancer risk: a review of the current genetic landscape'
- Author
-
Miriam J. Smith, D. Gareth Evans, Emma J Crosbie, Richard J. Edmondson, and Nicola Flaum
- Subjects
business.industry ,Cancer research ,Medicine ,Epithelial ovarian cancer ,Current (fluid) ,business - Published
- 2019
32. From process to progress-2017 International Conference on Neurofibromatosis 1, Neurofibromatosis 2 and Schwannomatosis
- Author
-
Eric Legius, Ludwine Messiaen, Miriam J. Smith, Aaron Schindeler, Scott R. Plotkin, D. Gareth Evans, Marco Giovannini, Karlyne M. Reilly, Rosalie E. Ferner, Annette Bakker, Nicole J. Ullrich, Larry S. Sherman, Ype Elgersma, Alison C. Lloyd, Brigitte C. Widemann, and Neurosciences
- Subjects
Multiple schwannomas ,Neurofibromatosis 2 ,medicine.medical_specialty ,Neurofibromatosis 1 ,Skin Neoplasms ,Neurofibromatoses ,Clinical Sciences ,Disease ,Article ,Neurofibromatosis ,Rare Diseases ,Genetics ,Animals ,Humans ,Medicine ,Schwannomatosis ,Genetics (clinical) ,Cancer ,Pediatric ,schwannomatosis ,business.industry ,Neurosciences ,Clinical course ,Multiple Neurofibromas ,medicine.disease ,Dermatology ,Clinical trial ,Disease Susceptibility ,business ,Neurilemmoma - Abstract
The neurofibromatoses are inherited, tumor suppressor disorders that are characterized by multiple, benign peripheral nerve sheath tumors and other nervous system tumors. Each disease is associated with a distinct genetic mutation and with a different pathogenesis and clinical course. Neurofibromatosis 1 (NF1) is common and epitomized by multiple neurofibromas with widespread complications. NF2 and schwannomatosis are rare diseases that are typified by multiple schwannomas that are particularly painful in people with schwannomatosis. Since 1985, the Children's Tumor Foundation (formerly the National Neurofibromatosis Foundation) has hosted an international Neurofibromatosis Conference, bringing together international participants who are focused on NF research and clinical care. The 2017 Conference, held in Washington, DC, was among the largest gatherings of NF researchers to date and included presentations from clinicians and basic scientists, highlighting new data regarding the molecular and cellular mechanisms underlying each of these diseases as well as results from clinical studies and clinical trials. This article summarizes the findings presented at the meeting and represents the current state-of-the art for NF research.
- Published
- 2019
33. Molecular Genetics of Meningioma
- Author
-
Miriam J. Smith
- Subjects
Genetics ,Meningioma ,medicine.medical_specialty ,business.industry ,Molecular genetics ,Mutation (genetic algorithm) ,Medicine ,Neurofibromatosis type 2 ,business ,medicine.disease - Published
- 2016
34. The Contribution of Whole Gene Deletions and Large Rearrangements to the Mutation Spectrum in Inherited Tumor Predisposing Syndromes
- Author
-
Helen Byers, Jill E. Urquhart, Andrew J Wallace, Miriam J. Smith, William G. Newman, Carolyn Gokhale, D. Gareth Evans, Naomi L. Bowers, Emma K. Miles, Elaine F. Harkness, and Michael Bulman
- Subjects
0301 basic medicine ,Genetics ,congenital, hereditary, and neonatal diseases and abnormalities ,Mutation ,Biology ,BRCA2 Protein ,MLH1 ,medicine.disease_cause ,Germline ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Germline mutation ,MSH2 ,030220 oncology & carcinogenesis ,Genotype ,medicine ,Chromosome 22 ,Genetics (clinical) - Abstract
Heterozygous whole gene deletions (WGDs), and intragenic microdeletions, account for a significant proportion of mutations underlying cancer predisposition syndromes. We analyzed the frequency and genotype-phenotype correlations of microdeletions in 12 genes (BRCA1, BRCA2, TP53, MSH2, MLH1, MSH6, PMS2, NF1, NF2, APC, PTCH1, and VHL) representing seven tumor predisposition syndromes in 5,897 individuals (2,611 families) from our center. Overall, microdeletions accounted for 14% of identified mutations. As expected, smaller deletions or duplications were more common (12%) than WGDs (2.2%). Where a WGD was identified in the germline in NF2, the mechanism of somatic second hit was not deletion, as previously described for NF1. For neurofibromatosis type 1 and 2, we compared the mechanism of germline deletion. Unlike NF1, where three specific deletion sizes account for most germline WGDs, NF2 deletion breakpoints were different across seven samples tested. One of these deletions was 3.93 Mb and conferred a severe phenotype, thus refining the region for a potential NF2 modifier gene to a 2.04-Mb region on chromosome 22. The milder phenotype of NF2 WGDs may be due to the apparent absence of chromosome 22 loss as the second hit. These observations of WGD phenotypes will be helpful for interpreting incidental findings from microarray analysis and next-generation sequencing.
- Published
- 2016
35. Abstract 2322: A polygenic risk score-based genetic stratification for endometrial cancer
- Author
-
Cemsel Bafligil, Miriam J. Smith, Artitaya Lophatananon, Deborah J. Thompson, Emma J Crosbie, Gareth D. Evans, and Joe Dennis
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Endometrial cancer ,Cancer ,Single-nucleotide polymorphism ,Genome-wide association study ,medicine.disease ,Internal medicine ,Epidemiology ,Cohort ,Genetic predisposition ,Medicine ,Polygenic risk score ,business - Abstract
Endometrial cancer (EC) is the most common of gynecological cancers in the UK. There is evidence for genetic predisposition to EC, however, single nucleotide polymorphisms have not been considered in risk prediction models so far. We hypothesized that a polygenic risk score (PRS) based on a panel of robust SNPs may be useful to predict women at high risk of developing EC. We conducted a systematic review to identify the most robust SNPs which may be associated with EC risk. The resulting panel of SNPs was validated in an independent cohort comprising EC cases, and control women with intact uterus. In the present study, we used 560 cases and 1202 controls of broad-European origin to construct a PRS. Our systematic review suggested twenty four SNPs, nineteen of which were reported to reach genome-wide significance in large GWAS. Preliminary results using 560 cases and 1202 controls produced effect sizes, directions and dosages similar to published data. The mean PRS for EC cases was 0.64 (SD=0.43, 95%CI 0.60-0.68) while mean PRS for controls was 0.48 (SD=0.42, 95% CI 0.46-0.51). The AUC of the PRS achieved was 0.61. Women in the highest tertile of the PRS score had a 2.4 times increased risk of developing EC compared to women in the lowest tertile (OR=2.40, 95%CI 1.85-3.09, p = 1.47E-11). Women in the mid tertile also had a modestly increased risk of EC compared to the lowest tertile (OR=1.40, 95%CI 1.06-1.84, p = 0.017). There was a clear trend of increasing risk with category (p = 6.25E-12). There is mounting evidence for polygenic input from common variants in EC susceptibility. Our results, for the first time, validate the potential use of a PRS for risk stratification of women at high risk of EC. When considered alongside epidemiological risk factors, the PRS could provide a good discrimination for early detection and prevention of EC in a clinical setting. Citation Format: Cemsel Bafligil, Deborah J. Thompson, Artitaya Lophatananon, Miriam J. Smith, Joe Dennis, Gareth D. Evans, Emma J. Crosbie. A polygenic risk score-based genetic stratification for endometrial cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 2322.
- Published
- 2020
36. An activation pathway governs cell wall polymerization by a bacterial morphogenic machine
- Author
-
Miriam J. Smith, Rohs Pda, Hansang Cho, Srisuknimit, Georgia R. Squyres, Andrew C. Kruse, Jackson Buss, Sim S, Emma R. Garner, Suzanne Walker, Daniel Kahne, Megan Sjodt, and Thomas G. Bernhardt
- Subjects
chemistry.chemical_classification ,0303 health sciences ,ATP synthase ,biology ,030306 microbiology ,Chemistry ,medicine.disease_cause ,MreB ,In vitro ,Cell wall ,03 medical and health sciences ,Enzyme ,Polymerization ,biology.protein ,medicine ,Biophysics ,sense organs ,Escherichia coli ,Actin ,030304 developmental biology - Abstract
Cell elongation in rod-shaped bacteria is mediated by the Rod system, a conserved morphogenic complex that spatially controls cell wall (CW) assembly. InEscherichia coli, alterations in a CW synthase component of the system called PBP2 were identified that overcome other inactivating defects. Rod system activity was stimulated in the suppressors in vivo, and purified synthase complexes with these changes showed more robust CW synthesis in vitro. Polymerization of the actin-like MreB component of the Rod system was also found to be enhanced in cells with the activated synthase. The results suggest an activation pathway governing Rod system function in which PBP2 conformation plays a central role in stimulating both CW glycan polymerization by its partner RodA and the formation of cytoskeletal filaments of MreB to orient CW assembly. An analogous activation pathway involving similar enzymatic components is likely responsible for controlling CW synthesis by the division machinery.
- Published
- 2018
37. Pain correlates with germline mutation in schwannomatosis
- Author
-
Michael E. Talkowski, James F. Gusella, Vanessa L. Merker, Justin T. Jordan, Gordon J. Harris, Vijaya Ramesh, Serkan Erdin, Alessandra Suuberg, Miriam A. Bredella, Miriam J. Smith, Scott R. Plotkin, James A. Walker, Marlon Seijo, and Wenli Cai
- Subjects
Male ,0301 basic medicine ,Skin Neoplasms ,SMARCB1 ,Gastroenterology ,Cohort Studies ,0302 clinical medicine ,Quality of life ,Whole Body Imaging ,pain ,Schwannomatosis ,Pain Measurement ,Medicine(all) ,medicine.diagnostic_test ,Visual Analog Pain Scale ,Cancer Pain ,SMARCB1 Protein ,General Medicine ,Middle Aged ,Magnetic Resonance Imaging ,Tumor Burden ,Peripheral ,Female ,Neurilemmoma ,Research Article ,Cohort study ,Adult ,medicine.medical_specialty ,Neurofibromatoses ,Observational Study ,03 medical and health sciences ,Germline mutation ,Internal medicine ,medicine ,Humans ,Genetic Association Studies ,Germ-Line Mutation ,schwannomatosis ,business.industry ,whole-body MRI ,Magnetic resonance imaging ,medicine.disease ,030104 developmental biology ,Quality of Life ,LZTR1 ,business ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Schwannomatosis has been linked to germline mutations in the SMARCB1 and LZTR1 genes, and is frequently associated with pain. In a cohort study, we assessed the mutation status of 37 patients with clinically diagnosed schwannomatosis and compared to clinical data, whole body MRI (WBMRI), visual analog pain scale, and Short Form 36 (SF-36) bodily pain subscale. We identified a germline mutation in LZTR1 in 5 patients (13.5%) and SMARCB1 in 15 patients (40.5%), but found no germline mutation in 17 patients (45.9%). Peripheral schwannomas were detected in 3 LZTR1-mutant (60%) and 10 SMARCB1-mutant subjects (66.7%). Among those with peripheral tumors, the median tumor number was 4 in the LZTR1 group (median total body tumor volume 30 cc) and 10 in the SMARCB1 group (median volume 85cc), (P=.2915 for tumor number and P = .2289 for volume). mutation was associated with an increased prevalence of spinal schwannomas (100% vs 41%, P = .0197). The median pain score was 3.9/10 in the LZTR1 group and 0.5/10 in the SMARCB1 group (P = .0414), and SF-36 pain-associated quality of life was significantly worse in the LZTR1 group (P = .0106). Pain scores correlated with total body tumor volume (rho = 0.32471, P = .0499), but not with number of tumors (rho = 0.23065, P = .1696). We found no significant difference in quantitative tumor burden between mutational groups, but spinal schwannomas were more common in LZTR1-mutant patients. Pain was significantly higher in LZTR1-mutant than in SMARCB1-mutant patients, though spinal tumor location did not significantly correlate with pain. This suggests a possible genetic association with schwannomatosis-associated pain.
- Published
- 2018
38. Inherited BRCA1 epimutation as a novel cause of breast and ovarian cancer
- Author
-
Jamie M Ellingford, Timothy J. Aitman, Glenda M. Beaman, Miriam J. Smith, Javier Santoyo-Lopez, William G. Newman, Andrew J Wallace, Helen Byers, Fiona Lalloo, D. Gareth Evans, Elke M van Veen, and Diana Eccles
- Subjects
endocrine system diseases ,Bisulfite sequencing ,Methylation ,Biology ,medicine.disease ,Germline ,DNA sequencing ,CpG site ,medicine ,Cancer research ,Allele ,Ovarian cancer ,skin and connective tissue diseases ,Gene - Abstract
BackgroundPathogenic variants in BRCA1 or BRCA2 are identified in ~20% of families with multiple individuals with early-onset breast/ovarian cancer. Extensive searches for additional highly penetrant genes or alternative mutational mechanisms altering BRCA1/2 have not explained the missing heritability. For the first time, we report transgenerational epigenetic silencing of BRCA1 due to promoter hypermethylation in two families with breast/ovarian cancer.MethodsBRCA1 promoter methylation of ten CpG dinucleotides in breast/ovarian cancer families without germline BRCA1/2 pathogenic variants was assessed by pyrosequencing and clonal bisulfite sequencing. RNA and DNA sequencing of BRCA1 from lymphocytes was undertaken to establish allelic expression and the presence of germline variants.FindingsBRCA1 promoter hypermethylation was identified in two of 49 families with multiple women affected with grade 3 breast/high grade serous ovarian cancer. Soma-wide BRCA1 promoter hypermethylation was confirmed in blood, buccal mucosa and hair follicles. Methylation levels were ~50%, consistent with the silencing of one allele and confirmed by clonal bisulfite sequencing. RNA sequencing revealed allelic loss of BRCA1 expression in both families and this segregated with a novel heterozygous variant c.-107A>T in the BRCA1 5’UTR.InterpretationOur results indicate a novel mechanism for familial breast/ovarian cancer, caused by epigenetic silencing of the BRCA1 promoter, segregating with an in cis 5’UTR variant in two independent families. We propose that methylation analyses are indicated in all families affected by early onset breast/ovarian cancer without a BRCA1/2 pathogenic variant.FundingFunded by Prevent Breast Cancer (GA 12-006 and GA 15-002) and the Manchester NIHR Biomedical Research Centre (IS-BRC-1215-20007).
- Published
- 2018
- Full Text
- View/download PDF
39. Sarcoma in neurofibromatosis 2:case report and review of the literature
- Author
-
Miriam J. Smith, Andrew J Wallace, D C Mangham, Anthony J. Freemont, M Bulman, D G R Evans, and C Linder
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Neurofibromatosis 2 ,Delayed Diagnosis ,030105 genetics & heredity ,Schwannoma ,Malignancy ,Diagnosis, Differential ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Fatal Outcome ,Immunochemistry ,Antineoplastic Combined Chemotherapy Protocols ,Genetics ,medicine ,otorhinolaryngologic diseases ,Humans ,Retroperitoneal Neoplasms ,SMARCB1 ,Neurofibromatosis type 2 ,Neurofibromatosis ,Genetics (clinical) ,Neurofibromin 2 ,Spinal Neoplasms ,business.industry ,Palliative Care ,Sarcoma ,SMARCB1 Protein ,Sorafenib ,medicine.disease ,Oncology ,Doxorubicin ,030220 oncology & carcinogenesis ,Neoplasm Grading ,Complication ,business - Abstract
Neurofibromatosis type 2 (NF2) is associated with the development of several types of benign nervous system tumours, while malignancies are rare. We report a 22-year-old man who presented with retroperitoneal and spinal high-grade sarcomas with epithelial features. Samples showed a mixed epithelioid and spindled cell content with little associated matrix and inconclusive immunochemistry. Genetic analysis of a schwannoma and matched blood samples demonstrated a constitutional de novo substitution at the splice donor site of intron 8 of the NF2 gene and aa acquired large deletion of the entire NF2 gene as a second hit, with some loss of SMARCB1. The sarcoma also showed evidence of loss of SMARCB1 and NF2 with loss of INI1 staining. Unfortunately the mass was unresectable and the patient died 6 months after diagnosis. This malignancy was most consistent with SMARCB1-deficient epithelioid malignant peripheral nerve sheath tumour, although a significant differential was proximal-type epithelial sarcoma. Each differential has previously been reported only once with NF2. This demonstrates an extremely rare potential complication of the condition.
- Published
- 2018
40. A deep intronic SMARCB1 variant associated with schwannomatosis
- Author
-
Meredith Wilson, Miriam J. Smith, Daffyd Gareth R. Evans, Francisco Cammarata-Scalisi, Catherine Banks, Lisa Ewans, Naomi L. Bowers, Andrew J Wallace, Jason Pinner, Rosanna Coates‐Brown, Sanjeev S. Bhaskar, and Katrina A. Morris
- Subjects
Adult ,Skin Neoplasms ,Neurofibromatoses ,Genetic Linkage ,business.industry ,MEDLINE ,SMARCB1 Protein ,Computational biology ,medicine.disease ,Introns ,Text mining ,Genetics ,Humans ,Medicine ,Female ,Genetic Predisposition to Disease ,SMARCB1 ,business ,Schwannomatosis ,Neurilemmoma ,Genetics (clinical) - Published
- 2019
41. SMARCE1 mutations in pediatric clear cell meningioma: case report
- Author
-
Linton T. Evans, Jack van Hoff, Miriam J. Smith, David F Bauer, William F. Hickey, D. Gareth Evans, and William G. Newman
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Chromosomal Proteins, Non-Histone ,Loss of Heterozygosity ,medicine.disease_cause ,Chromatin remodeling ,Resection ,Meningioma ,Meningeal Neoplasms ,medicine ,Clear Cell Meningioma ,Humans ,Spinal Cord Neoplasms ,Family history ,Allele ,Mutation ,Lumbar Vertebrae ,business.industry ,Laminectomy ,General Medicine ,medicine.disease ,SWI/SNF ,DNA-Binding Proteins ,Urinary Incontinence ,Child, Preschool ,business - Abstract
Clear cell meningioma (CCM) is an uncommon variant of meningioma. The authors describe a case of a pediatric CCM localized to the lumbar spine. After resection, sequencing revealed an inactivating mutation in the SWI/SNF chromatin remodeling complex subunit SMARCE1, with loss of the second allele in the tumor. The authors present a literature review of this mutation that is associated with CCM and a family history of spine tumors.
- Published
- 2015
42. Clinical and molecular predictors of mortality in neurofibromatosis 2: a UK national analysis of 1192 patients
- Author
-
D. Gareth Evans, Miriam J. Smith, Adam Shaw, Adrian L. Jones, Shazia K. Afridi, Lucy Raymond, Rupert Obholzer, Andrew T. King, Laura Heap, Jan M. Friedman, Allyson Parry, Patrick R. Axon, Amy Taylor, Dorothy Halliday, Andrew J Wallace, Adam Hexter, and Harry Joe
- Subjects
Adult ,Male ,Neurofibromatosis 2 ,medicine.medical_specialty ,Adolescent ,Kaplan-Meier Estimate ,medicine.disease_cause ,Genes, Neurofibromatosis 2 ,Internal medicine ,Epidemiology ,otorhinolaryngologic diseases ,Genetics ,medicine ,Humans ,Mutation type ,Missense mutation ,Neurofibromatosis ,Child ,Genetic Association Studies ,Genetics (clinical) ,Mutation ,Proportional hazards model ,business.industry ,Neurooncology ,Infant ,medicine.disease ,United Kingdom ,Child, Preschool ,Life expectancy ,Female ,business - Abstract
Background Neurofibromatosis 2 (NF2) is an autosomal-dominant tumour predisposition syndrome characterised by bilateral vestibular schwannomas, considerable morbidity and reduced life expectancy. Although genotype–phenotype correlations are well established in NF2, little is known about effects of mutation type or location within the gene on mortality. Improvements in NF2 diagnosis and management have occurred, but their effect on patient survival is unknown. Methods We evaluated clinical and molecular predictors of mortality in 1192 patients (771 with known causal mutations) identified through the UK National NF2 Registry. Kaplan–Meier survival and Cox regression analyses were used to evaluate predictors of mortality, with jackknife adjustment of parameter SEs to account for the strong intrafamilial phenotypic correlations that occur in NF2. Results The study included 241 deaths during 10 995 patient-years of follow-up since diagnosis. Early age at diagnosis and the presence of intracranial meningiomas were associated with increased mortality, and having a mosaic, rather than non-mosaic, NF2 mutation was associated with reduced mortality. Patients with splice-site or missense mutations had lower mortality than patients with truncating mutations (OR 0.459, 95% CI 0.213 to 0.990, and OR 0.196, 95% CI 0.213 to 0.990, respectively). Patients with splice-site mutations in exons 6–15 had lower mortality than patients with splice-site mutations in exons 1–5 (OR 0.333, 95% CI 0.129 to 0.858). The mortality of patients with NF2 diagnosed in more recent decades was lower than that of patients diagnosed earlier. Conclusions Continuing advances in molecular diagnosis, imaging and treatment of NF2-associated tumours offer hope for even better survival in the future.
- Published
- 2015
43. Germline Mutations in SUFU Cause Gorlin Syndrome–Associated Childhood Medulloblastoma and Redefine the Risk Associated With PTCH1 Mutations
- Author
-
Sanjeev S. Bhaskar, Sarah B. Daly, Simon G. Williams, William G. Newman, Jill E. Urquhart, Martin G. McCabe, Christian Beetz, James O'Sullivan, Beverley Anderson, Anna Kelsey, Deemesh Oudit, Miriam J. Smith, Edmund Cheesman, D. Gareth Evans, and Zaynab Bholah
- Subjects
Patched Receptors ,Risk ,Cancer Research ,Nonsense mutation ,Receptors, Cell Surface ,medicine.disease_cause ,Germline ,symbols.namesake ,Germline mutation ,medicine ,Humans ,Multiplex ligation-dependent probe amplification ,Cerebellar Neoplasms ,Germ-Line Mutation ,Exome sequencing ,Genetics ,Sanger sequencing ,Mutation ,business.industry ,Basal Cell Nevus Syndrome ,Magnetic Resonance Imaging ,Patched-1 Receptor ,Repressor Proteins ,stomatognathic diseases ,Oncology ,PTCH1 ,symbols ,business ,Medulloblastoma - Abstract
Purpose Heterozygous germline PTCH1 mutations are causative of Gorlin syndrome (naevoid basal cell carcinoma), but detection rates > 70% have rarely been reported. We aimed to define the causative mutations in individuals with Gorlin syndrome without PTCH1 mutations. Methods We undertook exome sequencing on lymphocyte DNA from four unrelated individuals from families with Gorlin syndrome with no PTCH1 mutations found by Sanger sequencing, multiplex ligation-dependent probe amplification (MLPA), or RNA analysis. Results A germline heterozygous nonsense mutation in SUFU was identified in one of four exomes. Sanger sequencing of SUFU in 23 additional PTCH1-negative Gorlin syndrome families identified a SUFU mutation in a second family. Copy-number analysis of SUFU by MLPA revealed a large heterozygous deletion in a third family. All three SUFU-positive families fulfilled diagnostic criteria for Gorlin syndrome, although none had odontogenic jaw keratocysts. Each SUFU-positive family included a single case of medulloblastoma, whereas only two (1.7%) of 115 individuals with Gorlin syndrome and a PTCH1 mutation developed medulloblastoma. Conclusion We demonstrate convincing evidence that SUFU mutations can cause classical Gorlin syndrome. Our study redefines the risk of medulloblastoma in Gorlin syndrome, dependent on the underlying causative gene. Previous reports have found a 5% risk of medulloblastoma in Gorlin syndrome. We found a < 2% risk in PTCH1 mutation–positive individuals, with a risk up to 20× higher in SUFU mutation–positive individuals. Our data suggest childhood brain magnetic resonance imaging surveillance is justified in SUFU-related, but not PTCH1-related, Gorlin syndrome.
- Published
- 2014
44. Mutations in LZTR1 add to the complex heterogeneity of schwannomatosis
- Author
-
Sarah B. Daly, Beverly Anderson, Simon G. Williams, James O'Sullivan, Wilfrid Richer, Miriam J. Smith, William G. Newman, Sanjeev S. Bhaskar, Daniel du Plessis, Amir Samii, Jill E. Urquhart, Christian Beetz, Samantha J Mills, Bertand Isidor, Cecilie F. Rustad, Dorothy Halliday, Sébastien Barbarot, Alan Fryer, D. Gareth Evans, and Franck Bourdeaut
- Subjects
Male ,Skin Neoplasms ,Neurofibromatoses ,Copy number analysis ,Loss of Heterozygosity ,Biology ,Article ,Germline ,Loss of heterozygosity ,symbols.namesake ,Germline mutation ,otorhinolaryngologic diseases ,medicine ,Humans ,Genetic Predisposition to Disease ,Schwannomatosis ,Germ-Line Mutation ,Exome sequencing ,Sanger sequencing ,Genetics ,Neuroma, Acoustic ,Sequence Analysis, DNA ,medicine.disease ,Pedigree ,Mutation (genetic algorithm) ,symbols ,Female ,Neurology (clinical) ,Neurilemmoma ,Transcription Factors - Abstract
Objectives: We aimed to determine the proportion of individuals in our schwannomatosis cohort whose disease is associated with an LZTR1 mutation. Methods: We used exome sequencing, Sanger sequencing, and copy number analysis to screen 65 unrelated individuals with schwannomatosis who were negative for a germline NF2 or SMARCB1 mutation. We also screened samples from 39 patients with a unilateral vestibular schwannoma (UVS), plus at least one other schwannoma, but who did not have an identifiable germline or mosaic NF2 mutation. Results: We identified germline LZTR1 mutations in 6 of 16 patients (37.5%) with schwannomatosis who had at least one affected relative, 11 of 49 (22%) sporadic patients, and 2 of 39 patients with UVS in our cohort. Three germline mutation–positive patients in total had developed a UVS. Mosaicism was excluded in 3 patients without germline mutation in NF2 , SMARCB1 , or LZTR1 by mutation screening in 2 tumors from each. Conclusions: Our data confirm the relationship between mutations in LZTR1 and schwannomatosis. They indicate that germline mutations in LZTR1 confer an increased risk of vestibular schwannoma, providing further overlap with NF2, and that further causative genes for schwannomatosis remain to be identified.
- Published
- 2014
45. Revisiting neurofibromatosis type 2 diagnostic criteria to exclude LZTR1-related schwannomatosis
- Author
-
Charlotte Hammerbeck-Ward, Michael Bulman, Simon R. Freeman, D. Gareth Evans, Scott A. Rutherford, Simon K L Lloyd, Carolyn Gokhale, Andrew J Wallace, Andrew T. King, Miriam J. Smith, and Naomi L. Bowers
- Subjects
0301 basic medicine ,Adult ,Male ,Pediatrics ,medicine.medical_specialty ,Neurofibromatosis 2 ,congenital, hereditary, and neonatal diseases and abnormalities ,Skin Neoplasms ,Databases, Factual ,Neurofibromatoses ,Loss of Heterozygosity ,Germline ,Functional Laterality ,Article ,Cohort Studies ,03 medical and health sciences ,Germline mutation ,0302 clinical medicine ,otorhinolaryngologic diseases ,Medicine ,Humans ,030212 general & internal medicine ,Neurofibromatosis type 2 ,Schwannomatosis ,Germ-Line Mutation ,Neurofibromin 2 ,business.industry ,Neuroma, Acoustic ,Middle Aged ,medicine.disease ,Dermatology ,030104 developmental biology ,Cohort ,Etiology ,Female ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Neurilemmoma ,Cohort study ,Transcription Factors - Abstract
Objective:To determine the specificity of the current clinical diagnostic criteria for neurofibromatosis type 2 (NF2) relative to the requirement for unilateral vestibular schwannoma (VS) and at least 2 other NF2-related tumors.Methods:We interrogated our Manchester NF2 database, which contained 205 individuals meeting NF2 criteria who initially presented with a unilateral VS. Of these, 83 (40.7%) went on to develop a contralateral VS. We concentrated our genetic analysis on a group of 70 who initially fulfilled NF2 criteria with a unilateral vestibular schwannoma and at least 2 additional nonintradermal schwannomas.Results:Overall, 5/70 (7%) individuals with unilateral VS and at least 2 other schwannomas had a pathogenic or likely pathogenic LZTR1 mutation. Twenty of the 70 subsequently developed bilateral disease. Of the remaining 50, 5 (10%) had a germline LZTR1 mutation, equivalent to the number (n = 5) with a germline NF2 mutation.Conclusions:The most common etiology for unilateral VS and 2 additional NF2-associated tumors in this cohort was mosaic NF2. Germline LZTR1 and germline NF2 mutations were equally common in our cohort. This indicates that LZTR1 must be considered when making a diagnosis of NF2 in the presence of unilateral VS in individuals without a germline NF2 mutation.
- Published
- 2017
46. Update from the 2013 international neurofibromatosis conference
- Author
-
Michel Kalamarides, Andrea I. McClatchey, Michael Fisher, Ludwine Messiaen, Thijs van der Vaart, Jaishri O. Blakeley, D. Gareth Evans, Alison C. Lloyd, Tena Rosser, Miriam J. Smith, David H. Gutmann, Helen Morrison, Dusica Babovic-Vuksanovic, Xandra O. Breakefield, Anne C. Albers, David A. Stevenson, Vincent M. Riccardi, Courtney M. Dunn, Catherine D. Van Raamsdonk, Aaron Schindeler, Jan M. Friedman, David Parkinson, Anat Stemmer-Rachamimov, Yuan Zhu, Annette Bakker, Marco Giovannini, Brian Weiss, Brigitte C. Widemann, Nicole J. Ullrich, and Scott R. Plotkin
- Subjects
Skin Neoplasms ,Neurofibromatoses ,Population ,Article ,Genetics ,Animals ,Humans ,Medicine ,Neurofibromatosis ,education ,Schwannomatosis ,Genetics (clinical) ,Clinical Trials as Topic ,education.field_of_study ,biology ,business.industry ,Research ,Congresses as Topic ,medicine.disease ,Neurofibromin 1 ,Chromosome 17 (human) ,Merlin (protein) ,Disease Models, Animal ,biology.protein ,business ,Chromosome 22 ,Neurilemmoma - Abstract
The 2013 Neurofibromatosis (NF) Conference took place at the Portola Hotel and Spa, Monterey, CA, from June 8-11, 2013. This international meeting is sponsored annually by the Children's Tumor Foundation (CTF), with the goal of bringing together NF researchers and clinicians from disparate fields of expertise. The conference agenda included a range of preclinical topics with a focus on signaling pathways and mouse models and a number of clinical topics, including a symposium on the interaction of academics, government, and industry in NF clinical trials. Neurofibromatosis is a group of inherited genetic disorders – NF1, NF2, and schwannomatosis – that together affect about 100,000 persons in the US. NF1 is the most common, with an estimated birth prevalence of 1:3,000 [Friedman., 1999; Lammert et al., 2005]; for NF2 and schwannomatosis, the estimated birth prevalence is 1:25,000 and 1:40,000, respectively [Antinheimo et al., 2000; Evans et al., 2005]. NF1, NF2 and schwannomatosis have in common that they predispose affected individuals to develop Schwann cell tumors such as neurofibromas and schwannomas. At a lower frequency, they also predispose to the development of a number of other benign and malignant tumor types, as well as some developmental abnormalities including learning disabilities (in NF1). The disorders arise from mutations in different genes, each of which plays a key role in regulating cellular function. The NF1 gene on human chromosome 17 encodes an intracellular signaling molecule that functions as a GTPase activating protein for Ras proteins, whereas the NF2 gene on human chromosome 22 encodes a cytoskeletal-membrane linking protein with reported roles in the suppression of several different growth-associated signaling pathways. The biology of schwannomatosis remains understudied despite the identification of germline SMARCB1 mutations in schwannomatosis patients in 2007.
- Published
- 2014
47. SMARCB1 mutations in schwannomatosis and genotype correlations with rhabdoid tumors
- Author
-
Miriam J. Smith, D. Gareth Evans, Naomi L. Bowers, Helen Eaton, and Andrew J Wallace
- Subjects
Proband ,Neurofibromatosis 2 ,Cancer Research ,Skin Neoplasms ,Neurofibromatoses ,Chromosomal Proteins, Non-Histone ,Biology ,medicine.disease_cause ,Germline ,Exon ,Genotype ,Genetics ,medicine ,Humans ,SMARCB1 ,Schwannomatosis ,Molecular Biology ,Genetic Association Studies ,Rhabdoid Tumor ,Genetic testing ,Neurofibromin 2 ,Mutation ,medicine.diagnostic_test ,Tumor Suppressor Proteins ,SMARCB1 Protein ,medicine.disease ,DNA-Binding Proteins ,Cancer research ,Neurilemmoma ,Transcription Factors - Abstract
Mutations in the SMARCB1 gene are involved in several human tumor-predisposing syndromes. They were established as an underlying cause of the tumor suppressor syndrome schwannomatosis in 2008. There is a much higher rate of mutation detection in familial disease than in sporadic disease. We have performed extensive genetic testing on a cohort of familial and sporadic patients who fulfilled clinical diagnostic criteria for schwannomatosis. In our updated cohort, we identified novel mutations within the SMARCB1 gene as well as several recurrent mutations. Of the schwannomatosis screens reported to date, including those in our updated cohort, SMARCB1 mutations have been found in 45% of familial probands and 9% of sporadic patients. The exon 1 mutation, c.41C>A p.Pro14His (10% in our series), and the 3' untranslated region mutation, c.*82C>T (27%), are the most common changes reported in patients with schwannomatosis to date, indicating the presence of mutation hot spots at both 5' and 3' portions of the gene. Comparison with germline SMARCB1 mutations in patients with rhabdoid tumors showed that the schwannomatosis mutations were significantly more likely to occur at either end of the gene and be nontruncating mutations (P < 0.0001). SMARCB1 mutations are found in a significant proportion of schwannomatosis patients, and an even higher proportion of rhabdoid patients. Whereas SMARCB1 alone seems to account for rhabdoid disease, there is likely to be substantial heterogeneity in schwannomatosis even for familial disease. There is a clear genotype-phenotype correlation, with germline rhabdoid mutations being significantly more likely to be centrally placed, involve multiple exon deletions, and be truncating mutations.
- Published
- 2014
48. Intronic splicing mutations in PTCH1 cause Gorlin syndrome
- Author
-
William G. Newman, D. Gareth Evans, Helen Byers, Miriam J. Smith, Zaynab Bholah, and Emma K. Miles
- Subjects
Male ,Patched Receptors ,Cancer Research ,Sequence analysis ,RNA Splicing ,DNA Mutational Analysis ,Receptors, Cell Surface ,Biology ,medicine.disease_cause ,Frameshift mutation ,symbols.namesake ,Genetics ,medicine ,Humans ,Multiplex ligation-dependent probe amplification ,Genetics (clinical) ,Sanger sequencing ,Mutation ,Base Sequence ,Intron ,Basal Cell Nevus Syndrome ,Molecular biology ,Introns ,Patched-1 Receptor ,genomic DNA ,Oncology ,RNA splicing ,symbols ,Female ,RNA Splice Sites ,Multiplex Polymerase Chain Reaction - Abstract
Gorlin syndrome is an autosomal dominant disorder characterized by multiple early-onset basal cell carcinoma, odontogenic keratocysts and skeletal abnormalities. It is caused by heterozygous mutations in the tumour suppressor PTCH1. Routine clinical genetic testing, by Sanger sequencing and multiplex ligation-dependent probe amplification (MLPA) to confirm a clinical diagnosis of Gorlin syndrome, identifies a mutation in 60–90 % of cases. We undertook RNA analysis on lymphocytes from ten individuals diagnosed with Gorlin syndrome, but without known PTCH1 mutations by exonic sequencing or MLPA. Two altered PTCH1 transcripts were identified. Genomic DNA sequence analysis identified an intron 7 mutation c.1068-10T>A, which created a strong cryptic splice acceptor site, leading to an intronic insertion of eight bases; this is predicted to create a frameshift p.(His358Alafs*12). Secondly, a deep intronic mutation c.2561-2057A>G caused an inframe insertion of 78 intronic bases in the cDNA transcript, leading to a premature stop codon p.(Gly854fs*3). The mutations are predicted to cause loss of function of PTCH1, consistent with its tumour suppressor function. The findings indicate the importance of RNA analysis to detect intronic mutations in PTCH1 not identified by routine screening techniques.
- Published
- 2014
49. Update from the 2011 International Schwannomatosis Workshop: From genetics to diagnostic criteria
- Author
-
Larry S. Sherman, Jaishri O. Blakeley, Kim Hunter-Schaedle, D. Gareth Evans, Jeremie Vitte, Laura Papi, Ludwine Messiaen, Miriam J. Smith, Scott R. Plotkin, Theo J. M. Hulsebos, Marco Giovannini, C. Oliver Hanemann, Ganjam V. Kalpana, Bruce R. Korf, Nancy Ratner, and Anat Stemmer-Rachamimov
- Subjects
Genetics ,Biology ,medicine.disease ,Bioinformatics ,Article ,Germline ,Germline mutation ,medicine ,Allele ,SMARCB1 ,Neurofibromatosis ,Schwannomatosis ,Chromosome 22 ,Genetics (clinical) ,Neurofibromatoses - Abstract
Schwannomatosis is the third major form of neurofibromatosis and is characterized by the development of multiple schwannomas in the absence of bilateral vestibular schwannomas. The 2011 Schwannomatosis Update was organized by the Children’s Tumor Foundation (www.ctf.org) and held in Los Angeles, CA, from June 5–8, 2011. This article summarizes the highlights presented at the Conference and represents the “state-of-the-field” in 2011. Genetic studies indicate that constitutional mutations in the SMARCB1 tumor suppressor gene occur in 40–50% of familial cases and in 8–10% of sporadic cases of schwannomatosis. Tumorigenesis is thought to occur through a four-hit, three-step model, beginning with a germline mutation in SMARCB1 (hit 1), followed by loss of a portion of chromosome 22 that contains the second SMARCB1 allele and one NF2 allele (hits 2 and 3), followed by mutation of the remaining wild-type NF2 allele (hit 4). Insights from research on HIV and pediatric rhabdoid tumors have shed light on potential molecular pathways that are dysregulated in schwannomatosis-related schwannomas. Mouse models of schwannomatosis have been developed and promise to further expand our understanding of tumorigenesis and the tumor microenvironment. Clinical reports have described the occurrence of intracranial meningiomas in schwannomatosis patients and in families with germline SMARCB1 mutations. The authors propose updated diagnostic criteria to incorporate new clinical and genetic findings since 2005. In the next 5 years, the authors expect that advances in basic research in the pathogenesis of schwannomatosis will lead toward clinical investigations of potential drug therapies.
- Published
- 2013
50. Genetic Predisposition in Young Patients with Solitary Meningiomas
- Author
-
John-Paul Kilday, Ian Kamaly-Asl, Omar N. Pathmanaban, Andy King, Miriam J. Smith, and D G R Evans
- Subjects
Pathology ,medicine.medical_specialty ,business.industry ,Genetic predisposition ,Medicine ,Neurology (clinical) ,business ,Bioinformatics - Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.