Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Eur J Hum Genet ; 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117932

ABSTRACT

Rhabdoid tumor predisposition syndrome type-1 (RTPS1) is characterized by germline pathogenic variants in SMARCB1 and development of INI1-deficient rhabdoid tumors in early childhood. Due to its poor prognosis, the risk of subsequent tumor development and the impact of surveillance at later ages are poorly understood. We retrospectively reviewed individuals referred to the Cancer Genetics Program at The Hospital for Sick Children for SMARCB1 genetic testing and/or surveillance for RTPS1. In addition, to explore characteristics of late-onset tumors in RTPS1, a literature review was conducted. Of eighty-three individuals (55 probands and 28 family members), 12 probands and 4 family members were genetically confirmed with RTPS1. Four pediatric probands with RTPS1 underwent surveillance. An additional three individuals, including one patient with 22q11.2 distal deletion without history of tumor, one patient with negative genetic testing results but clinically diagnosed with RTPS1, and one sibling identified through cascade testing, underwent surveillance. Three patients with RTPS1 developed tumors between the ages of 9 and 17, including malignant rhabdoid tumors (N = 3), schwannomas (N = 4), and epithelioid malignant peripheral nerve sheath tumor (N = 1). Three of these lesions were asymptomatically detected by surveillance. A literature review revealed 17 individuals with RTPS1 who developed INI1-deficient tumors after age five. Individuals with RTPS1 remain at elevated risk for developing INI1-deficient tumors after the peak age of rhabdoid tumor in early childhood. Extension of surveillance beyond 5 years of age could lead to improved survival and reduced morbidity for these patients, and prospective evaluation of revised approaches will be important.

2.
Pediatr Blood Cancer ; 68(8): e29021, 2021 08.
Article in English | MEDLINE | ID: mdl-33788392

ABSTRACT

PURPOSE: Individuals with cancer predisposition syndromes (CPS) are often followed in cancer screening programs, which aim to detect early stage tumors. While cancer surveillance has the potential to improve patient outcomes, its psychosocial impact is uncharacterized in the pediatric population. We examined the cancer surveillance experience from the perspectives of adolescents and parents of children at risk of developing cancer. PATIENTS AND METHODS: Using grounded theory and thematic analysis qualitative methodology, we conducted semi-structured interviews with parents and adolescents, separately. Interviews were transcribed verbatim and coded separately to derive overlapping and unique themes. RESULTS: We completed 20 semi-structured interviews (11 parents and nine adolescents). Positive experiences were related to feelings of reassurance and taking a proactive approach. Both adolescents and parents experienced worry, related to practical aspects of screening, and related to the reminder of cancer risk that manifests with surveillance appointments. This worry was cyclical, associated with appointments, and generally waned over time. Participants felt that the benefits of surveillance outweighed perceived challenges. Open communication with health care providers, and equipping parents/adolescents with vocabulary to discuss their diagnosis and care with others, were felt to be important for mitigating worries associated with cancer risk and surveillance. CONCLUSION: Parents and adolescents experience worry associated with surveillance for CPS, which may warrant regular psychosocial support, particularly during the first year following CPS diagnosis. Enhancing communication with the health care team and among and beyond immediate family members represents an additional important strategy to mitigate adverse experiences and perceptions.


Subject(s)
Caregivers , Disease Susceptibility , Neoplasms , Adolescent , Child , Communication , Humans , Neoplasms/diagnosis , Parents , Qualitative Research
3.
Clin Genet ; 96(5): 461-467, 2019 11.
Article in English | MEDLINE | ID: mdl-31368132

ABSTRACT

Von Hippel-Lindau disease (VHL) is a heritable condition caused by pathogenic variants in VHL and is characterized by benign and malignant lesions in the central nervous system (CNS) and abdominal viscera. Due to its variable expressivity, existing efforts to collate VHL patient data do not adequately capture all VHL manifestations. We developed a comprehensive and standardized VHL database in the web-based application, REDCap, that thoroughly captures all VHL manifestation data. As an initial trial, information from 86 VHL patients from the University Health Network/Hospital for Sick Children was populated into the database. Analysis of this cohort showed missense variants occurring with the greatest frequency, with all variants localizing to the α- or ß-domains of VHL. The most prevalent manifestations were central nervous system (CNS), renal, and retinal neoplasms, which were associated with frameshift variants and large deletions. We observed greater age-related penetrance for CNS hemangioblastomas with truncating variants compared to missense, while the reverse was true for pheochromocytomas. We demonstrate the utility of a comprehensive VHL database, which supports the standardized collection of clinical and genetic data specific to this patient population. Importantly, we expect that its web-based design will facilitate broader international collaboration and lead to a better understanding of VHL.


Subject(s)
Hemangioblastoma/genetics , Pheochromocytoma/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , von Hippel-Lindau Disease/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Canada/epidemiology , Central Nervous System/metabolism , Central Nervous System/pathology , Child , Child, Preschool , Female , Hemangioblastoma/epidemiology , Hemangioblastoma/pathology , Humans , Male , Middle Aged , Mutation, Missense/genetics , Pedigree , Penetrance , Pheochromocytoma/epidemiology , Pheochromocytoma/pathology , Young Adult , von Hippel-Lindau Disease/epidemiology , von Hippel-Lindau Disease/pathology
4.
Pediatr Blood Cancer ; 66(1): e27445, 2019 01.
Article in English | MEDLINE | ID: mdl-30207072

ABSTRACT

BACKGROUND: Hereditary tumor predisposition syndromes (HTPSs) are being recognized more frequently in the etiology of pediatric cancer. Previous research indicates that disclosure of tumor susceptibility is a significant event in adolescents' lives. Insight into adolescents' adjustment to knowledge of their syndromes can guide healthcare delivery, particularly genetic counseling. This study explored the experiences of adolescents with hereditary tumor predisposition and their perceptions of living at risk. METHODS: Seven adolescents, ages 14 to 17, representing six different childhood-onset HTPSs, were purposively sampled and interviewed using a study-specific semistructured interview guide. We explored the disclosure process, support systems, and the perceived benefits and harms of knowledge of hereditary tumor susceptibility. Interview transcripts were analyzed via interpretive description. RESULTS: Three major themes emerged from the data: (1) The benefits of knowledge outweigh the harms; (2) context surrounding genetic testing must be recognized; and (3) self-concept is influenced but not defined by tumor risk. CONCLUSIONS: We conclude that adolescents recognize the challenges associated with awareness of tumor predisposition but may also identify positive aspects in their experiences, reflecting a changed life perspective. Results of this exploratory study suggest strategies that can guide pretest and posttest genetic counseling of adolescents for HTPSs, facilitating the adaptive incorporation of genetic information into an adolescent's self-concept.


Subject(s)
Genetic Counseling , Genetic Predisposition to Disease , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/psychology , Adolescent , Female , Follow-Up Studies , Genetic Testing , Humans , Male , Qualitative Research , Surveys and Questionnaires
6.
J Med Genet ; 55(6): 395-402, 2018 06.
Article in English | MEDLINE | ID: mdl-29437867

ABSTRACT

BACKGROUND: von Hippel-Lindau (vHL) syndrome is a rare autosomal-dominant disorder that confers a lifelong risk for developing both benign and malignant tumours in multiple organs. Recent evidence suggests that vHL may exhibit genetic anticipation (GA). The aim of this study was to determine if GA occurs in vHL, and if telomere shortening may be a factor in GA. METHODS: A retrospective chart review of vHL families seen at The Hospital for Sick Children between 1984 and 2016 was performed. Age of onset (AOO, defined as the age of first physician-diagnosed vHL-related manifestation) was confirmed for 96 patients from 20 unrelated families (80 clinically affected and 16 unaffected carriers). Flow-FISH(flow cytometry sorting of cells whose telomeres are labeled by Fluorescence In Situ Hybridization) was used to measure mean telomere length of six white blood cell subtypes from 14 known VHL pathogenic variant carriers. RESULTS: The median AOO for generations I, II and III were 32.5, 22.5 and 12.0 years, respectively. The differences in the AOO between generations were highly significant using a Cox proportional hazards model (P=6.00×10-12). Telomere lengths were significantly different for granulocytes and natural killer lymphocytes of patients with vHL compared with age-matched controls. For six vHL parent-child pairs, median white blood cell telomere lengths between parent and child were not significantly different. CONCLUSIONS: Our results suggest that vHL telomere abnormalities may be primarily somatic in origin rather than a cause of GA. As tumour development exhibits GA in our cohort, vHL surveillance guidelines may need to account for a patient's generational position within a vHL pedigree.


Subject(s)
Anticipation, Genetic , Genetic Predisposition to Disease , Telomere Shortening/genetics , von Hippel-Lindau Disease/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Flow Cytometry , Granulocytes/metabolism , Granulocytes/pathology , Heterozygote , Humans , In Situ Hybridization, Fluorescence , Infant , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Male , Pedigree , Telomere/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Young Adult , von Hippel-Lindau Disease/pathology
7.
Pediatr Blood Cancer ; 65(1)2018 Jan.
Article in English | MEDLINE | ID: mdl-28960912

ABSTRACT

BACKGROUND: To expand the current knowledge of DICER1 syndrome and to propose criteria for genetic testing based on experience at a pediatric tertiary care center. PROCEDURE: This study involved a retrospective chart review of the 78 patients (47 probands and 31 family members) seen in the Cancer Genetics Program at The Hospital for Sick Children (SickKids) who were offered genetic testing for DICER1. RESULTS: Of 47 probands offered genetic testing for DICER1, 46 pursued testing: 11 (23.9%) carried a pathogenic variant and one proband (2.1%) carried a missense variant of uncertain significance with evidence for pathogenicity. Thirty-one family members of variant-positive probands were offered testing: eight of the 25 who agreed to testing carried their familial variant (32.0%). Overall, 20 patients were identified to have a variant in DICER1 (eight males, 12 females). Of these, 13 (65.0%) presented with clinical manifestations associated with the syndrome. The most common lesions were pleuropulmonary blastoma (PPB) (five of 20 patients, 25.0%) and pineoblastoma (three of 20 patients, 15.0%). The average age at which individuals were diagnosed with a primary neoplasm was 5.2 years (range 0.8-20 years, median 3.0). Surveillance at our institution, with a median follow-up time of 23 months, has identified PPB in two asymptomatic individuals. These lesions were identified at early stages, thus potentially reducing treatment-related morbidity and mortality. CONCLUSION: This study further delineates the DICER1 syndrome phenotype and demonstrates the feasibility of a DICER1 syndrome surveillance protocol for the early detection of tumors.


Subject(s)
Brain Neoplasms/genetics , Neoplastic Syndromes, Hereditary/genetics , Pineal Gland , Pinealoma/genetics , Pulmonary Blastoma/genetics , Adolescent , Adult , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Neoplastic Syndromes, Hereditary/mortality , Neoplastic Syndromes, Hereditary/pathology , Pinealoma/mortality , Pinealoma/pathology , Pulmonary Blastoma/mortality , Pulmonary Blastoma/pathology
8.
Clin Cancer Res ; 23(13): e91-e97, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28674117

ABSTRACT

As the understanding of the genetic etiology of childhood cancers increases, the need for the involvement of experts familiar with the provision of genetic counseling for this population is paramount. In October 2016, the American Association for Cancer Research organized the AACR Childhood Cancer Predisposition Workshop in which international experts in pediatric cancer predisposition met to establish surveillance guidelines for children with cancer predisposition. Identifying for whom, when, why, and how these cancer predisposition surveillance guidelines should be implemented is essential. Genetic counselors invited to this workshop provide a genetic counseling framework for oncology professionals in this article. Points of entry and recommendations regarding the provision and timing of the initial and subsequent genetic counseling sessions are addressed. The genetic counseling and testing processes are reviewed, and the psychologic impact related to surveillance is explored. Pediatric cancer genetics will continue to grow and evolve as a field, and genetic counseling services will be vital to ensure appropriate identification and management of at-risk children moving forward. Clin Cancer Res; 23(13); e91-e97. ©2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.


Subject(s)
Genetic Counseling/trends , Genetic Predisposition to Disease/epidemiology , Medical Oncology/trends , Neoplasms/diagnosis , Child , Counselors , Genetic Testing/trends , Humans , Neoplasms/epidemiology , Neoplasms/genetics , Pediatrics/trends , Risk Assessment
9.
Clin Cancer Res ; 23(13): e107-e114, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28674119

ABSTRACT

Hereditary gastrointestinal cancer predisposition syndromes have been well characterized, but management strategies and surveillance remain a major challenge, especially in childhood. In October 2016, the American Association for Cancer Research organized the AACR Childhood Cancer Predisposition Workshop in which international experts in care of children with a hereditary risk of cancer met to define surveillance strategies and management of children with cancer predisposition syndromes. In this article, we review the current literature in polyposis syndromes that can be diagnosed in childhood and may be associated with an increased incidence of gastrointestinal neoplasms and other cancer types. These disorders include adenomatous polyposis syndromes (APC and MUTYH), juvenile polyposis coli (BMPR1A and SMAD4), Peutz-Jeghers Syndrome (STK11/LKB1), and PTEN hamartoma tumor syndrome (PHTS; PTEN), which can present with a more limited juvenile polyposis phenotype. Herein, the panel of experts provides recommendations for clinical diagnosis, approach to genetic testing, and focus on cancer surveillance recommendations when appropriate during the pediatric period. We also review current controversies on genetic evaluation of patients with hepatoblastoma and indications for surveillance for this tumor. Childhood cancer risks and surveillance associated with disorders involving the mismatch repair genes, including Lynch syndrome and constitutional mismatch repair deficiency (CMMRD), are discussed elsewhere in this series. Clin Cancer Res; 23(13); e107-e14. ©2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.


Subject(s)
Adenomatous Polyposis Coli/genetics , Gastrointestinal Neoplasms/genetics , Hamartoma Syndrome, Multiple/genetics , Peutz-Jeghers Syndrome/genetics , Adenomatous Polyposis Coli/diagnosis , Adenomatous Polyposis Coli/epidemiology , Child , Early Detection of Cancer , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/epidemiology , Genetic Predisposition to Disease , Genetic Testing , Hamartoma Syndrome, Multiple/diagnosis , Hamartoma Syndrome, Multiple/epidemiology , Humans , Pediatrics , Peutz-Jeghers Syndrome/diagnosis , Peutz-Jeghers Syndrome/epidemiology
10.
Clin Cancer Res ; 23(13): e123-e132, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28674121

ABSTRACT

Children and adolescents who present with neuroendocrine tumors are at extremely high likelihood of having an underlying germline predisposition for the multiple endocrine neoplasia (MEN) syndromes, including MEN1, MEN2A and MEN2B, MEN4, and hyperparathyroid-jaw tumor (HPT-JT) syndromes. Each of these autosomal dominant syndromes results from a specific germline mutation in unique genes: MEN1 is due to pathogenic MEN1 variants (11q13), MEN2A and MEN2B are due to pathogenic RET variants (10q11.21), MEN4 is due to pathogenic CDKN1B variants (12p13.1), and the HPT-JT syndrome is due to pathogenic CDC73 variants (1q25). Although each of these genetic syndromes share the presence of neuroendocrine tumors, each syndrome has a slightly different tumor spectrum with specific surveillance recommendations based upon tumor penetrance, including the age and location for which specific tumor types most commonly present. Although the recommended surveillance strategies for each syndrome contain similar approaches, important differences do exist among them. Therefore, it is important for caregivers of children and adolescents with these syndromes to become familiar with the unique diagnostic criteria for each syndrome, and also to be aware of the specific tumor screening and prophylactic surgery recommendations for each syndrome. Clin Cancer Res; 23(13); e123-e32. ©2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.


Subject(s)
Adenoma/genetics , Fibroma/genetics , Hyperparathyroidism/genetics , Jaw Neoplasms/genetics , Multiple Endocrine Neoplasia/genetics , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Adenoma/diagnosis , Adenoma/epidemiology , Adolescent , Child , Fibroma/diagnosis , Fibroma/epidemiology , Genetic Predisposition to Disease , Germ-Line Mutation/genetics , Humans , Hyperparathyroidism/diagnosis , Hyperparathyroidism/epidemiology , Jaw Neoplasms/diagnosis , Jaw Neoplasms/epidemiology , Multiple Endocrine Neoplasia/diagnosis , Multiple Endocrine Neoplasia/epidemiology , Multiple Endocrine Neoplasia Type 2b/diagnosis , Multiple Endocrine Neoplasia Type 2b/epidemiology , Multiple Endocrine Neoplasia Type 2b/genetics , Risk Factors
11.
Clin Cancer Res ; 23(12): e54-e61, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28620005

ABSTRACT

The neurofibromatoses consist of at least three autosomal-dominant inherited disorders: neurofibromatosis type 1 (NF1), neurofibromatosis type 2 (NF2), and schwannomatosis. For over 80 years, these conditions were inextricably tied together under generalized neurofibromatosis. In 1987, the localization of NF1 to chromosome 17q and NF2 (bilateral vestibular schwannoma) to 22q led to a consensus conference at Bethesda, Maryland. The two main neurofibromatoses, NF1 and NF2, were formally separated. More recently, the SMARCB1 and LZTR1 genes on 22q have been confirmed as causing a subset of schwannomatosis. The last 26 years have seen a great improvement in understanding of the clinical and molecular features of these conditions as well as insights into management. Childhood presentation of NF2 (often with meningioma) in particular predicts a severe multitumor disease course. Malignancy is rare in NF2, particularly in childhood; however, there are substantial risks from benign and low-grade central nervous system (CNS) tumors necessitating MRI surveillance to optimize management. At least annual brain MRI, including high-resolution images through the auditory meatus, and a clinical examination and auditory assessment are required from diagnosis or from around 10 to 12 years of age if asymptomatic. Spinal imaging at baseline and every 2 to 3 years is advised with more frequent imaging if warranted on the basis of sites of tumor involvement. The malignancy risk in schwannomatosis is not well defined but may include an increased risk of malignant peripheral nerve sheath tumor in SMARCB1 Imaging protocols are also proposed for SMARCB1 and LZTR1 schwannomatosis and SMARCE1-related meningioma predisposition. Clin Cancer Res; 23(12); e54-e61. ©2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Meningioma/genetics , Neurilemmoma/genetics , Neurofibromatoses/genetics , SMARCB1 Protein/genetics , Skin Neoplasms/genetics , Transcription Factors/genetics , Adolescent , Child , Genetic Predisposition to Disease , Humans , Magnetic Resonance Imaging , Meningioma/diagnosis , Meningioma/diagnostic imaging , Meningioma/epidemiology , Neurilemmoma/diagnosis , Neurilemmoma/diagnostic imaging , Neurilemmoma/epidemiology , Neurofibromatoses/diagnosis , Neurofibromatoses/diagnostic imaging , Neurofibromatoses/epidemiology , Neurofibromin 1/genetics , Neurofibromin 2/genetics , Skin Neoplasms/diagnosis , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/epidemiology
12.
Clin Cancer Res ; 23(12): e76-e82, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28620008

ABSTRACT

PTEN hamartoma tumor syndrome (PHTS), DICER1 syndrome, and hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome are pleiotropic tumor predisposition syndromes that include benign and malignant neoplasms affecting adults and children. PHTS includes several disorders with shared and distinct clinical features. These are associated with elevated lifetime risk of breast, thyroid, endometrial, colorectal, and renal cancers as well as melanoma. Thyroid cancer represents the predominant cancer risk under age 20 years. DICER1 syndrome includes risk for pleuropulmonary blastoma, cystic nephroma, ovarian sex cord-stromal tumors, and multinodular goiter and thyroid carcinoma as well as brain tumors including pineoblastoma and pituitary blastoma. Individuals with HLRCC may develop multiple cutaneous and uterine leiomyomas, and they have an elevated risk of renal cell carcinoma. For each of these syndromes, a summary of the key syndromic features is provided, the underlying genetic events are discussed, and specific screening is recommended. Clin Cancer Res; 23(12); e76-e82. ©2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.


Subject(s)
DEAD-box RNA Helicases/genetics , Fumarate Hydratase/genetics , Hamartoma Syndrome, Multiple/genetics , Leiomyomatosis/genetics , Neoplastic Syndromes, Hereditary/genetics , PTEN Phosphohydrolase/genetics , Ribonuclease III/genetics , Skin Neoplasms/genetics , Uterine Neoplasms/genetics , Child , Early Detection of Cancer , Hamartoma Syndrome, Multiple/epidemiology , Hamartoma Syndrome, Multiple/pathology , Humans , Leiomyomatosis/epidemiology , Leiomyomatosis/pathology , Neoplastic Syndromes, Hereditary/epidemiology , Neoplastic Syndromes, Hereditary/pathology , Risk Factors , Skin Neoplasms/epidemiology , Skin Neoplasms/pathology , Uterine Neoplasms/epidemiology , Uterine Neoplasms/pathology
13.
Clin Cancer Res ; 23(12): e68-e75, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28620007

ABSTRACT

Von Hippel-Lindau disease (vHL) is a hereditary tumor predisposition syndrome that places affected individuals at risk for multiple tumors, which are predominantly benign and generally occur in the central nervous system or abdomen. Although the majority of tumors occur in adults, children and adolescents with the condition develop a significant proportion of vHL manifestations and are vulnerable to delayed tumor detection and their sequelae. Although multiple tumor screening paradigms are currently being utilized for patients with vHL, surveillance should be reassessed as the available relevant clinical information continues to expand. We propose a new vHL screening paradigm similar to existing approaches, with important modifications for some tumor types, placing an emphasis on risks in childhood. This includes advancement in the timing of surveillance initiation and increased frequency of screening evaluations. Another neuroendocrine-related familial condition is the rapidly expanding hereditary paraganglioma and pheochromocytoma syndrome (HPP). The tumor spectrum for patients with HPP syndrome includes paragangliomas, pheochromocytomas, renal cancer, and gastrointestinal stromal tumors. The majority of patients with HPP syndrome harbor an underlying variant in one of the SHDx genes (SDHA, SDHB, SDHC, SDHD, SDHA, and SDHAF2), although other genes also have been described (MAX and TMEM127). Annual screening for elevated plasma or urine markers along with complete blood count and biennial whole-body MRI accompanied by focal neck MRI is recommended for older children and adults with HPP syndrome to detect tumors early and to decrease morbidity and mortality from HPP-related tumors. Clin Cancer Res; 23(12); e68-e75. ©2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.


Subject(s)
Adrenal Gland Neoplasms/diagnosis , Paraganglioma/diagnosis , Pheochromocytoma/diagnosis , von Hippel-Lindau Disease/diagnosis , Adrenal Gland Neoplasms/epidemiology , Adrenal Gland Neoplasms/pathology , Early Detection of Cancer , Humans , Mutation , Neoplasm Proteins/genetics , Paraganglioma/epidemiology , Paraganglioma/genetics , Paraganglioma/pathology , Pheochromocytoma/epidemiology , Pheochromocytoma/genetics , Pheochromocytoma/pathology , Risk Factors , von Hippel-Lindau Disease/epidemiology , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/pathology
14.
Pediatr Radiol ; 47(10): 1292-1301, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28474256

ABSTRACT

BACKGROUND: DICER1 syndrome, arising from a mutation in the DICER1 gene mapped to chromosome 14q32, is associated with an increased risk of a range of benign and malignant neoplasms. OBJECTIVE: To determine the spectrum of abnormalities and imaging characteristics in patients with DICER1 syndrome at a tertiary pediatric hospital. MATERIALS AND METHODS: This retrospective analysis evaluated imaging in patients ≤18 years with DICER1 germline variants between January 2004 and July 2016. An imaging database search including keywords pleuropulmonary blastoma, cystic nephroma, pineoblastoma, embryonal rhabdomyosarcoma, ovarian sex cord-stromal tumor, ovarian Sertoli-Leydig cell tumor and DICER1 syndrome, was cross-referenced against the institutional Cancer Genetics Program database, excluding patients with negative/unknown DICER1 gene testing. RESULTS: Sixteen patients were included (12 females; mean age at presentation: 4.2 years, range: 14 days to 17 years), with surveillance imaging encompassing the following modalities: chest X-ray and CT; abdominal, pelvic and neck US; and brain and whole-body MRI. Malignant lesions (68.8% of patients) included pleuropulmonary blastoma (5), pineoblastoma (3), ovarian Sertoli-Leydig cell tumor (1), embryonal rhabdomyosarcoma (1) and renal sarcoma (1); benign lesions (37.5% of patients) included thyroid cysts (2), thyroid nodules (2), cystic nephroma (2), renal cysts (1) and pineal cyst (1). A common lesional appearance observed across modalities and organs was defined as the "cracked windshield" sign. CONCLUSION: The spectrum of DICER1-related tumors and the young age at presentation suggest early surveillance of at-risk patients is critical, while minimizing exposure to ionizing radiation.


Subject(s)
DEAD-box RNA Helicases/genetics , Neoplasms/diagnostic imaging , Neoplasms/genetics , Ribonuclease III/genetics , Adolescent , Child , Child, Preschool , Diagnosis, Differential , Female , Genetic Predisposition to Disease , Humans , Infant , Infant, Newborn , Male , Mutation , Phenotype , Retrospective Studies , Syndrome
15.
Lancet Oncol ; 17(9): 1295-305, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27501770

ABSTRACT

BACKGROUND: Carriers of a germline TP53 pathogenic variant have a substantial lifetime risk of developing cancer. In 2011, we did a prospective observational study of members of families who chose to either undergo a comprehensive surveillance protocol for individuals with Li-Fraumeni syndrome or not. We sought to update our assessment of and modify the surveillance protocol, so in this study we report both longer follow-up of these patients and additional patients who underwent surveillance, as well as update the originally presented surveillance protocol. METHODS: A clinical surveillance protocol using physical examination and frequent biochemical and imaging studies (consisting of whole-body MRI, brain MRI, breast MRI, mammography, abdominal and pelvic ultrasound, and colonoscopy) was introduced at three tertiary care centres in Canada and the USA on Jan 1, 2004, for carriers of TP53 pathogenic variants. After confirmation of TP53 mutation, participants either chose to undergo surveillance or chose not to undergo surveillance. Patients could cross over between groups at any time. The primary outcome measure was detection of asymptomatic tumours by surveillance investigations. The secondary outcome measure was 5 year overall survival established from a tumour diagnosed symptomatically (in the non-surveillance group) versus one diagnosed by surveillance. We completed survival analyses using an as-treated approach. FINDINGS: Between Jan 1, 2004, and July 1, 2015, we identified 89 carriers of TP53 pathogenic variants in 39 unrelated families, of whom 40 (45%) agreed to surveillance and 49 (55%) declined surveillance. 19 (21%) patients crossed over from the non-surveillance to the surveillance group, giving a total of 59 (66%) individuals undergoing surveillance for a median of 32 months (IQR 12-87). 40 asymptomatic tumours have been detected in 19 (32%) of 59 patients who underwent surveillance. Two additional cancers were diagnosed between surveillance assessments (false negatives) and two biopsied lesions were non-neoplastic entities on pathological review (false positives). Among the 49 individuals who initially declined surveillance, 61 symptomatic tumours were diagnosed in 43 (88%) patients. 21 (49%) of the 43 individuals not on surveillance who developed cancer were alive compared with 16 (84%) of the 19 individuals undergoing surveillance who developed cancer (p=0·012) after a median follow-up of 46 months (IQR 22-72) for those not on surveillance and 38 months (12-86) for those on surveillance. 5 year overall survival was 88·8% (95% CI 78·7-100) in the surveillance group and 59·6% (47·2-75·2) in the non-surveillance group (p=0·0132). INTERPRETATION: Our findings show that long-term compliance with a comprehensive surveillance protocol for early tumour detection in individuals with pathogenic TP53 variants is feasible and that early tumour detection through surveillance is associated with improved long-term survival. Incorporation of this approach into clinical management of these patients should be considered. FUNDING: Canadian Institutes for Heath Research, Canadian Cancer Society, Terry Fox Research Institute, SickKids Foundation, and Soccer for Hope Foundation.


Subject(s)
Biomarkers, Tumor/metabolism , Germ-Line Mutation/genetics , Li-Fraumeni Syndrome/genetics , Multimodal Imaging/methods , Neoplasms/genetics , Population Surveillance , Tumor Suppressor Protein p53/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Follow-Up Studies , Genetic Predisposition to Disease , Heterozygote , Humans , Infant , Infant, Newborn , Li-Fraumeni Syndrome/diagnostic imaging , Li-Fraumeni Syndrome/metabolism , Li-Fraumeni Syndrome/pathology , Male , Middle Aged , Neoplasm Staging , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Neoplasms/pathology , Prognosis , Prospective Studies , Survival Rate , Young Adult
17.
Pediatr Blood Cancer ; 63(7): 1272-5, 2016 07.
Article in English | MEDLINE | ID: mdl-26928971

ABSTRACT

Anaplastic sarcoma of kidney (ASK) is a rare neoplasm recently associated with DICER1 mutations. We report a child with germline DICER1 mutation who developed ASK in preexisting septated renal cysts, which were likely cystic nephroma. From age 2.5 to 6 years, sonographic imaging illustrated changes in the size and number of renal cysts, followed at age 8.8 years by a mass, pathologically an ASK. Lung cysts resected in infancy were diagnosed retrospectively as pleuropulmonary blastoma. Both tumors had acquired somatic DICER1 mutations. Ultrasonographic evolution of renal cysts to ASK has not previously been documented. Children with both pulmonary and renal cysts are candidates for DICER1 mutation testing.


Subject(s)
Cysts , DEAD-box RNA Helicases/genetics , Genetic Diseases, Inborn , Kidney Neoplasms , Pulmonary Blastoma , Ribonuclease III/genetics , Sarcoma , Child , Child, Preschool , Cysts/genetics , Cysts/pathology , Cysts/surgery , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Genetic Diseases, Inborn/surgery , Humans , Infant , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Pulmonary Blastoma/genetics , Pulmonary Blastoma/pathology , Pulmonary Blastoma/surgery , Sarcoma/genetics , Sarcoma/pathology , Sarcoma/surgery , Syndrome
18.
Am J Gastroenterol ; 111(2): 275-84, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26729549

ABSTRACT

OBJECTIVES: Hereditary biallelic mismatch repair deficiency (BMMRD) is caused by biallelic mutations in the mismatch repair (MMR) genes and manifests features of neurofibromatosis type 1, gastrointestinal (GI) polyposis, and GI, brain, and hematological cancers. This is the first study to characterize the GI phenotype in BMMRD using both retrospective and prospective surveillance data. METHODS: The International BMMRD Consortium was created to collect information on BMMRD families referred from around the world. All patients had germline biallelic MMR mutations or lack of MMR protein staining in normal and tumor tissue. GI screening data were obtained through medical records with annual updates. RESULTS: Thirty-five individuals from seven countries were identified with BMMRD. GI data were available on 24 of 33 individuals (73%) of screening age, totaling 53 person-years. The youngest age of colonic adenomas was 7, and small bowel adenoma was 11. Eight patients had 19 colorectal adenocarcinomas (CRC; median age 16.7 years, range 8-25), and 11 of 18 (61%) CRC were distal to the splenic flexure. Eleven patients had 15 colorectal surgeries (median 14 years, range 9-25). Four patients had five small bowel adenocarcinomas (SBC; median 18 years, range 11-33). Two CRC and two SBC were detected during surveillance within 6-11 months and 9-16 months, respectively, of last consecutive endoscopy. No patient undergoing surveillance died of a GI malignancy. Familial clustering of GI cancer was observed. CONCLUSIONS: The prevalence and penetrance of GI neoplasia in children with BMMRD is high, with rapid development of carcinoma. Colorectal and small bowel surveillance should commence at ages 3-5 and 8 years, respectively.


Subject(s)
Adenocarcinoma/surgery , Adenoma/surgery , Brain Neoplasms/physiopathology , Colorectal Neoplasms/surgery , Intestine, Small/surgery , Neoplastic Syndromes, Hereditary/physiopathology , Adaptor Proteins, Signal Transducing/genetics , Adenocarcinoma/etiology , Adenocarcinoma/genetics , Adenoma/etiology , Adenoma/genetics , Adenosine Triphosphatases/genetics , Adolescent , Adult , Alleles , Brain Neoplasms/complications , Brain Neoplasms/etiology , Brain Neoplasms/genetics , Child , Child, Preschool , Colorectal Neoplasms/complications , Colorectal Neoplasms/etiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/physiopathology , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Female , Germ-Line Mutation , Glioma/etiology , Humans , Intestinal Neoplasms/etiology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/surgery , Kidney Neoplasms/etiology , Leukemia/etiology , Lymphoma/etiology , Male , Melanoma/etiology , Mismatch Repair Endonuclease PMS2 , MutL Protein Homolog 1 , Neoplastic Syndromes, Hereditary/complications , Neoplastic Syndromes, Hereditary/genetics , Nuclear Proteins/genetics , Phenotype , Prospective Studies , Retrospective Studies , Wilms Tumor/etiology , Young Adult
19.
Hum Pathol ; 46(6): 917-22, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25836323

ABSTRACT

Embryonal rhabdomyosarcoma (ERMS), a soft tissue sarcoma, is one of the most common pediatric cancers. Certain ERMSs are associated with the DICER1 syndrome, a tumor predisposition syndrome caused by germ-line DICER1 mutations. Characteristic somatic mutations have also been identified in DICER1-associated tumor types. These "hotspot" mutations affect the catalytic activity of the DICER1 ribonuclease IIIb domain. Primary ovarian ERMS (oERMS) is extremely rare. We present a case of a 6-year-old girl with an oERMS harboring 2 DICER1 mutations. The girl also exhibited other DICER1 phenotypes: cystic nephroma (CN) and multinodular goiter. Somatic investigations of the CN identified a hotspot DICER1 mutation different from that in the oERMS. Significantly, the CN presented at 12 years of age, which is much older than the previously reported age range of susceptibility. This report documents the occurrence of DICER1 mutations in a case of oERMS, expanding the spectrum of DICER1-associated tumors.


Subject(s)
DEAD-box RNA Helicases/genetics , Mutation/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Rhabdomyosarcoma, Embryonal/genetics , Rhabdomyosarcoma, Embryonal/pathology , Ribonuclease III/genetics , Child , DEAD-box RNA Helicases/metabolism , Female , Humans , MicroRNAs/genetics , Ovarian Neoplasms/diagnosis , Phenotype , Rhabdomyosarcoma, Embryonal/diagnosis , Ribonuclease III/metabolism
20.
Nat Genet ; 47(3): 257-62, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25642631

ABSTRACT

DNA replication-associated mutations are repaired by two components: polymerase proofreading and mismatch repair. The mutation consequences of disruption to both repair components in humans are not well studied. We sequenced cancer genomes from children with inherited biallelic mismatch repair deficiency (bMMRD). High-grade bMMRD brain tumors exhibited massive numbers of substitution mutations (>250/Mb), which was greater than all childhood and most cancers (>7,000 analyzed). All ultra-hypermutated bMMRD cancers acquired early somatic driver mutations in DNA polymerase ɛ or δ. The ensuing mutation signatures and numbers are unique and diagnostic of childhood germ-line bMMRD (P < 10(-13)). Sequential tumor biopsy analysis revealed that bMMRD/polymerase-mutant cancers rapidly amass an excess of simultaneous mutations (∼600 mutations/cell division), reaching but not exceeding ∼20,000 exonic mutations in <6 months. This implies a threshold compatible with cancer-cell survival. We suggest a new mechanism of cancer progression in which mutations develop in a rapid burst after ablation of replication repair.


Subject(s)
Base Pair Mismatch , Brain Neoplasms/genetics , DNA Mismatch Repair , DNA Replication/genetics , DNA Repair , DNA-Directed DNA Polymerase/genetics , Exons , Germ-Line Mutation , Humans , Microsatellite Instability
SELECTION OF CITATIONS
SEARCH DETAIL