Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters

Country/Region as subject
Publication year range
2.
Am J Med Genet A ; 194(1): 64-69, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37705207

ABSTRACT

Turner syndrome (45,X) is caused by a complete or partial absence of a single X chromosome. Vascular malformations occur due to abnormal development of blood and/or lymphatic vessels. They arise from either somatic or germline pathogenic variants in the genes regulating growth and apoptosis of vascular channels. Aortic abnormalities are a common, known vascular anomaly of Turner syndrome. However, previous studies have described other vascular malformations as a rare feature of Turner syndrome and suggested that vascular abnormalities in individuals with Turner syndrome may be more generalized. In this study, we describe two individuals with co-occurrence of Turner syndrome and vascular malformations with a lymphatic component. In these individuals, genetic testing of the lesional tissue revealed a somatic pathogenic variant in PIK3CA-a known and common cause of lymphatic malformations. Based on this finding, we conclude that the vascular malformations presented here and likely those previously in the literature are not a rare part of the clinical spectrum of Turner syndrome, but rather a separate clinical entity that may or may not co-occur in individuals with Turner syndrome.


Subject(s)
Cardiovascular Abnormalities , Lymphatic Abnormalities , Turner Syndrome , Vascular Malformations , Humans , Turner Syndrome/complications , Turner Syndrome/genetics , Mosaicism , Lymphatic Abnormalities/genetics , Vascular Malformations/complications , Vascular Malformations/genetics , Class I Phosphatidylinositol 3-Kinases/genetics
3.
Pediatr Dev Pathol ; 27(1): 3-12, 2024.
Article in English | MEDLINE | ID: mdl-37771132

ABSTRACT

BACKGROUND: Next generation sequencing (NGS) has increased the detection of fusion genes in cancer. NGS has found multiple fusions in single tumor samples; however, the incidence of this in pediatric soft tissue and bone tumors (PSTBTs) is not well documented. The aim of this study is to catalogue the incidence of multiple fusions in a series of PSTBTs, and apply a modified gene fusion classification system to determine clinical relevance. METHODOLOGY: RNA from 78 bone and soft tissue tumors and 7 external quality assessment samples were sequenced and analyzed using recently-described Metafusion (MF) software and classified using a modification of previously-published schema for fusion classification into 3 tiers: 1, strong clinical significance; 2, potential clinical significance; and 3, unknown clinical significance. RESULTS: One-hundred forty-five fusions were detected in 85 samples. Fifty-five samples (65%) had a single fusion and 30 (35%) had more than 1 fusion. No samples contained more than 1 tier 1 fusion. There were 40 tier 1 (28%), 36 tier 2 (24%), and 69 (48%) tier 3 fusions. CONCLUSIONS: A significant percentage of PSTBTs harbor more than 1 fusion, and by applying a modified fusion classification scheme, the potential clinical relevance of such fusions can be determined.


Subject(s)
Bone Neoplasms , Soft Tissue Neoplasms , Humans , Child , Incidence , Bone Neoplasms/genetics , Soft Tissue Neoplasms/genetics , Gene Fusion , Base Sequence , High-Throughput Nucleotide Sequencing , Oncogene Proteins, Fusion/genetics
4.
Cancer ; 129(14): 2245-2255, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37081608

ABSTRACT

BACKGROUND: Inhibition of the WEE1 kinase by adavosertib (AZD1775) potentiates replicative stress from genomic instability or chemotherapy. This study reports the pediatric solid tumor phase 2 results of the ADVL1312 trial combining irinotecan and adavosertib. METHODS: Pediatric patients with recurrent neuroblastoma (part B), medulloblastoma/central nervous system embryonal tumors (part C), or rhabdomyosarcoma (part D) were treated with irinotecan and adavosertib orally for 5 days every 21 days. The combination was considered effective if there were at least three of 20 responses in parts B and D or six of 19 responses in part C. Tumor tissue was analyzed for alternative lengthening of telomeres and ATRX. Patient's prior tumor genomic analyses were provided. RESULTS: The 20 patients with neuroblastoma (part B) had a median of three prior regimens and 95% had a history of prior irinotecan. There were three objective responses (9, 11, and 18 cycles) meeting the protocol defined efficacy end point. Two of the three patients with objective responses had tumors with alternative lengthening of telomeres. One patient with pineoblastoma had a partial response (11 cycles), but parts C and D did not meet the protocol defined efficacy end point. The combination was well tolerated and there were no dose limiting toxicities at cycle 1 or beyond in any parts of ADVL1312 at the recommended phase 2 dose. CONCLUSION: This is first phase 2 clinical trial of adavosertib in pediatrics and the first with irinotecan. The combination may be of sufficient activity to consider further study of adavosertib in neuroblastoma.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Neuroblastoma , Rhabdomyosarcoma , Child , Humans , Irinotecan/therapeutic use , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Rhabdomyosarcoma/drug therapy , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/etiology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Protein-Tyrosine Kinases , Cell Cycle Proteins
5.
Histopathology ; 82(6): 946-952, 2023 May.
Article in English | MEDLINE | ID: mdl-36648026

ABSTRACT

AIMS: Ossifying fibromyxoid tumor (OFMT) is a rare enigmatic tumor of uncertain differentiation that can be classified as typical, atypical, and malignant subtypes based on cellularity, nuclear grade, and mitotic activity. The majority of OFMTs, regardless of the risk of malignancy, harbor genetic translocations. We report two malignant OFMTs, including one with evidence of dedifferentiation, with novel genefusions. METHODS AND RESULTS: Case 1 was a 63-year-old male with a dedifferentiated OFMT arising in the right wrist, while case 2 was a 41-year-old male with a malignant OFMT presenting as a posterior mediastinal mass. Case 2 showed multifocal expression with EMA and synaptophysin, while desmin and S100 were absent in both tumors. NGS sequencing studies detected PHF1::FOXR1 and PHF1::FOXR2 gene fusions in cases 1 and 2, respectively. Despite aggressive regimens, both progressed with wide spread metastases resulting in death within six years of diagnosis. CONCLUSIONS: We expand the genetic spectrum of OFMTs with two novel gene fusions, PHF1::FOXR1 and PHF1::FOXR2. These cases confirm the previously reported tendencies for OFMTs with rare variant fusions to demonstrate malignant behavior, unusual morphology, and non-specific immunophenotype.


Subject(s)
Fibroma, Ossifying , Fibroma , Soft Tissue Neoplasms , Male , Humans , Middle Aged , Adult , Fibroma, Ossifying/pathology , Soft Tissue Neoplasms/pathology , Fibroma/pathology , Gene Fusion , DNA-Binding Proteins/genetics , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Forkhead Transcription Factors/genetics
6.
Pediatr Blood Cancer ; : e30419, 2023 May 17.
Article in English | MEDLINE | ID: mdl-37194624

ABSTRACT

Complex lymphatic anomalies are debilitating conditions characterized by aberrant development of the lymphatic vasculature (lymphangiogenesis). Diagnosis is typically made by history, examination, radiology, and histologic findings. However, there is significant overlap between conditions, making accurate diagnosis difficult. Recently, genetic analysis has been offered as an additional diagnostic modality. Here, we describe four cases of complex lymphatic anomalies, all with PIK3CA variants but with varying clinical phenotypes. Identification of PIK3CA resulted in transition to a targeted inhibitor, alpelisib. These cases highlight the genetic overlap between phenotypically diverse lymphatic anomalies.

7.
Histopathology ; 80(1): 4-18, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34958503

ABSTRACT

Kinase alterations are increasingly recognised as oncogenic drivers in mesenchymal tumours. Infantile fibrosarcoma and the related renal tumour, congenital mesoblastic nephroma, were among the first solid tumours shown to harbour recurrent tyrosine kinase fusions, with the canonical ETV6::NTRK3 fusion identified more than 20 years ago. Although targeted testing has long been used in diagnosis, the advent of more robust sequencing techniques has driven the discovery of kinase alterations in an array of mesenchymal tumours. As our ability to identify these genetic alterations has improved, as has our recognition and understanding of the tumours that harbour these alterations. Specifically, this study will focus upon mesenchymal tumours harbouring NTRK or other kinase alterations, including tumours with an infantile fibrosarcoma-like appearance, spindle cell tumours resembling lipofibromatosis or peripheral nerve sheath tumours and those occurring in adults with a fibrosarcoma-like appearance. As publications describing the histology of these tumours increase so, too, do the variety kinase alterations reported, now including NTRK1/2/3, RET, MET, RAF1, BRAF, ALK, EGFR and ABL1 fusions or alterations. To date, these tumours appear locally aggressive and rarely metastatic, without a clear link between traditional features used in histological grading (e.g. mitotic activity, necrosis) and outcome. However, most of these tumours are amenable to new targeted therapies, making their recognition of both diagnostic and therapeutic import. The goal of this study is to review the clinicopathological features of tumours with NTRK and other tyrosine kinase alterations, discuss the most common differential diagnoses and provide recommendations for molecular confirmation with associated treatment implications.


Subject(s)
Fibrosarcoma/genetics , Gene Rearrangement , Neoplasms, Connective and Soft Tissue/genetics , Nephroma, Mesoblastic/genetics , Receptor, trkA/genetics , Fibrosarcoma/pathology , Humans , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasms, Connective and Soft Tissue/pathology , Nephroma, Mesoblastic/pathology
8.
Pediatr Dev Pathol ; 25(6): 668-671, 2022.
Article in English | MEDLINE | ID: mdl-36262073

ABSTRACT

Hepatocellular adenomas (HCA) in infants are exceedingly rare with only 5 cases reported to the best of our knowledge, all of them preceding the classification of HCA. Here we present an autopsy case of a 9-month-old girl with Burn-McKeown syndrome with an incidental liver nodule in the right lobe measuring 1.5 cm in greatest dimension. The lesion was composed of an unencapsulated proliferation of hepatocytes with multiple unaccompanied arteries without well-formed portal tracts, and an intact reticulin framework without thickened hepatic plates, findings consistent with an HCA. Glutamine synthetase (GS), lipid fatty acid-binding protein (LFABP), c-reactive protein (CRP), serum amyloid-a (SAA), beta-catenin and CD34 immunostains were performed. GS was diffusely and strongly positive in the lesion, CD34 showed heterogenous staining of sinusoids within the lesion without a well-formed rim from the background liver and beta-catenin was negative for nuclear staining. CRP and SAA were considered negative, and LFABP was retained. Molecular testing showed no CTNNB1 variants and found two tier 3 variants involving CHEK2 and PTEN genes. These findings are consistent with an unclassified HCA (U-HCA) per the 2019 WHO Classification of Tumors, representing the youngest patient reported. This raises the possibility that some HCAs are congenital or develop very early in life, remaining undiagnosed until later in life.


Subject(s)
Adenoma, Liver Cell , Carcinoma, Hepatocellular , Liver Neoplasms , Female , Infant , Humans , Adenoma, Liver Cell/diagnosis , Adenoma, Liver Cell/genetics , Adenoma, Liver Cell/pathology , beta Catenin/genetics , Carcinoma, Hepatocellular/diagnosis , Liver Neoplasms/genetics , Immunohistochemistry , Biomarkers, Tumor/genetics , Glutamate-Ammonia Ligase/metabolism , Fatty Acid-Binding Proteins/metabolism
9.
Am J Dermatopathol ; 44(1): 54-57, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34291746

ABSTRACT

ABSTRACT: In the past decade, there have been major advances in knowledge related to mesenchymal tumors, and new genetic alterations are being delineated. We report a mesenchymal spindle cell neoplasm harboring a novel gene fusion in an infant. Histopathologically, the neoplasm shared some features with sclerosing perineurioma, but immunohistochemically, EMA was negative, whereas GLUT1, NK1-C3, and BCOR were positive. Next-generation sequencing revealed a PCMTD1-pleomorphic adenoma gene 1 (PLAG1) fusion. PLAG1 contributes to the expression of a variety of genes implicated in regulating cell proliferation, and PCMTD1 has been related to the development of certain carcinomas. Recently, other soft tissue tumors in young children associated with PLAG1 fusion variants have been reported. Perhaps, mesenchymal neoplasms presenting PLAG1 fusions with different genes would confirm a specific group (PLAG mesenchymal tumours or "plagomas") in the near future.


Subject(s)
Chondrosarcoma, Mesenchymal/genetics , Soft Tissue Neoplasms/genetics , Chondrosarcoma, Mesenchymal/diagnosis , DNA-Binding Proteins , Finger Joint/physiopathology , Gene Fusion , Humans , Infant , Male , Protein D-Aspartate-L-Isoaspartate Methyltransferase , Soft Tissue Neoplasms/diagnosis
10.
Genes Chromosomes Cancer ; 60(9): 631-634, 2021 09.
Article in English | MEDLINE | ID: mdl-33840146

ABSTRACT

Ossifying fibromyxoid tumor (OFMT) is a rare soft tissue neoplasm of uncertain differentiation that has the capacity for local recurrence and metastasis. Many OFMTs, including typical, atypical, and malignant tumors, have demonstrated recurrent gene fusions. The fusion partners reported to date share a common core function in that they play either a direct or indirect role in processes influencing histone modification. Herein, we report an OFMT with unusual morphology and non-specific immunoprofile harboring a novel MEAF6-SUZ12 fusion. A 34-year-old male presented with a slowly growing mass in the right antecubital fossa. Excision demonstrated a 6.9 cm partially encapsulated, tan-white, lobulated, and calcified lesion. Microscopic evaluation demonstrated cytologically bland spindle to ovoid cells arranged in a haphazard manner within a fibromyxoid background containing dense collagen, often with sclerotic nodules, and randomly distributed ossification. The tumor cells were diffusely positive for CD34 while essentially negative for S100, desmin, MUC4, SOX10, AE1/3, SMA, and EMA. Next-generation sequencing studies (sarcoma gene fusion next-generation sequencing panel with subsequent Sanger confirmation) performed on formalin-fixed paraffin-embedded tissue detected a fusion product between MEAF6 exon 4 (NM_001270875) and SUZ12 exon 2 (NM_001321207.1). The proposed mechanism of pathogenesis in OFMT, namely epigenetic dysregulation, is reinforced by the fact that both of these partner genes are involved in histone modification.


Subject(s)
Fibroma/genetics , Histone Acetyltransferases/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Soft Tissue Neoplasms/genetics , Transcription Factors/genetics , Adult , Fibroma/pathology , Humans , Male , Ossification, Heterotopic/genetics , Ossification, Heterotopic/pathology , Soft Tissue Neoplasms/pathology
11.
Pediatr Blood Cancer ; 68(6): e28933, 2021 06.
Article in English | MEDLINE | ID: mdl-33565241

ABSTRACT

Pediatric histiocytic neoplasms are hematopoietic disorders frequently driven by the BRAF-V600E mutation. Here, we identified two BRAF gene fusions (novel MTAP-BRAF and MS4A6A-BRAF) in two aggressive histiocytic neoplasms. In contrast to previously described BRAF fusions, MTAP-BRAF and MS4A6A-BRAF do not respond to the paradox breaker RAF inhibitor (RAFi) PLX8394 due to stable fusion dimerization mediated by the N-terminal fusion partners. This highlights a significant and clinically relevant shift from the current dogma that BRAF-fusions respond similarly to BRAF-inhibitors. As an alternative, we show suppression of fusion-driven oncogenic growth with the pan-RAFi LY3009120 and MEK inhibition.


Subject(s)
Histiocytosis , Neoplasms , Cell Line, Tumor , Child , Humans , Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics
12.
Genes Chromosomes Cancer ; 59(8): 495-499, 2020 08.
Article in English | MEDLINE | ID: mdl-32222087

ABSTRACT

A novel group of S100- and CD34-positive spindle cell tumors with distinctive stromal and perivascular hyalinization harboring recurrent gene fusions involving kinases including RAF1, BRAF, NTRK1/2/3, and RET have been recently reported. To our knowledge, no such cases harboring ALK rearrangements have been identified. We report a previously healthy 41-year-old male with a 12-cm intramuscular shoulder mass. The tumor was composed of bland-appearing spindled to epithelioid cells, arranged in a patternless pattern in a background of loose myxoid stroma containing striking amianthoid-like stromal collagen and perivascular rings. In accordance with the previously reported tumors, the tumor cells showed diffuse immunopositivity with S100 and CD34, while lacking SOX10 expression. Targeted RNA-based next-generation sequencing identified a novel serine/threonine-protein phosphatase PP1-beta-catalytic subunit (PPP1CB)-ALK fusion gene. Although ALK break-apart was not detected by FISH, likely due to a paracentric inversion of chromosome 2, the presence of the fusion was confirmed by Sanger sequencing showing a 10-bp linker between exon 6 of PPP1CB and intron 19 of ALK while maintaining reading frame. Subsequent ALK-1 immunostain exhibited diffuse cytoplasmic staining in the tumor cells. Our case expands the molecular genetic spectrum of the distinctive group of spindle cell tumors with CD34/S100+ immunophenotype, supporting the important role of various kinases as drivers of oncogenesis. Awareness of this entity including its unique morphologic and immunophenotypic features as well as its interchangeable kinase gene fusions is crucial for correct classification and potential targeted therapy, particularly in aggressive subsets.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Oncogene Proteins, Fusion/genetics , Perivascular Epithelioid Cell Neoplasms/genetics , Protein Phosphatase 1/genetics , Adult , Antigens, CD34/genetics , Antigens, CD34/metabolism , Blood Vessels/pathology , Humans , Male , Perivascular Epithelioid Cell Neoplasms/pathology , S100 Proteins/genetics , S100 Proteins/metabolism , Shoulder/pathology , Stromal Cells/pathology
13.
Mod Pathol ; 33(4): 576-590, 2020 04.
Article in English | MEDLINE | ID: mdl-31690781

ABSTRACT

Inflammatory myofibroblastic tumors arising in infants are rare, poorly investigated and mostly reported as isolated cases or as a part of larger series thus, their clinicopathological and molecular features are essentially unknown. Archival files from two large pediatric institutions and a tumor registry were queried for pediatric inflammatory myofibroblastic tumors. Available material from patients ≤12 months of age was reviewed. Additional immunostains (ALK-1, D240, WT1) and ALK-FISH studies were performed as needed. Targeted anchored multiplex PCR with next-generation sequencing was done in all cases. A total of 12 of 131 infantile cases (mean 5.5 months) were identified (M:F of 2:1). Anatomic locations included intestinal/mesenteric (n = 6), head/neck (n = 3), and viscera (n = 3). Half of tumors showed a hypocellular myxoid pattern, perivascular condensation, and prominent vasculature with vague glomeruloid structures present in four of them. The remaining cases exhibited a more cellular pattern with minimal myxoid component. ALK-1 immunohistochemistry was positive in most cases (11/12) with cytoplasmic-diffuse (n = 6), cytoplasmic-granular (n = 2), and dot-like (n = 3) staining patterns. ALK fusion partners identified in five cases included EML4, TPM4, RANBP2, and a novel KLC1. Three inflammatory myofibroblastic tumors showed fusions with other kinases including TFG-ROS1 and novel FN1-ROS1 and RBPMS-NTRK3 rearrangements. Favorable outcome was documented in most cases (10/11) with available follow-up (median 17 months) while three patients were successfully treated with crizotinib. In summary, infantile inflammatory myofibroblastic tumors are rare and can exhibit paucicellular, extensively myxoid/vascular morphology with peculiar immunophenotype mimicking other mesenchymal or vascular lesions. All tumors harbored kinase fusions involving ALK, ROS1, and NTRK3 including three novel fusion partners (KLC1, FN1, and RBPMS, respectively). A favorable response to crizotinib seen in three cases supports its potential use in infants as seen in older patients. Awareness of these unusual morphologic, immunophenotypic, and molecular features is critical for appropriate diagnosis and optimized targeted therapy.


Subject(s)
Biomarkers, Tumor/genetics , Myofibroblasts/pathology , Neoplasms, Muscle Tissue/genetics , Neoplasms, Muscle Tissue/pathology , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/pathology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Crizotinib/therapeutic use , Female , Gene Fusion , Gene Rearrangement , Genetic Predisposition to Disease , Humans , Infant , Infant, Newborn , Italy , Kinesins , Male , Myofibroblasts/drug effects , Myofibroblasts/enzymology , Neoplasms, Muscle Tissue/drug therapy , Neoplasms, Muscle Tissue/enzymology , Phenotype , Philadelphia , Protein Kinase Inhibitors/therapeutic use , Registries , Soft Tissue Neoplasms/drug therapy , Soft Tissue Neoplasms/enzymology
14.
Histopathology ; 76(7): 1032-1041, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31994201

ABSTRACT

AIMS: The classification of paediatric spindle mesenchymal tumours is evolving, and the spectrum of so-called 'infantile fibrosarcoma' has expanded to include tumours with NTRK, BRAF and MET gene fusions. RET-rearranged paediatric spindle cell neoplasms are an emerging group; there is sparse literature on their clinical, pathological and genetic features, and their nosological place in the canon of soft tissue tumours is uncertain. In this study, we report five RET-rearranged paediatric spindle cell tumours with fusion partners MYH10, KIAA1217 and CLIP2. METHODS AND RESULTS: The tumours occurred in the pelvic region, paraspinal region, kidney and subcutaneous tissue of hand and abdomen. The patients' ages ranged from 6 months to 13 years (median 1 year). The tumours were composed of monomorphic spindle cells arranged in a fascicular pattern. Lesional cells had minimally atypical ovoid or tapered nuclei and pale cytoplasm with indistinct borders. Necrosis was not identified. Mitoses numbered three to 12 per 10 high-power field. Cases showed inconsistent and variable expression of S100, CD34 and SMA. Clinical behaviour ranged from small lesions potentially cured by simple resection to large lesions exhibiting metastasis, but responsive to kinase inhibitor therapy. CONCLUSIONS: Our findings help to define RET-rearranged spindle cell tumours. Although it is likely that these tumours comprise part of the morphological and clinical spectrum of infantile fibrosarcoma (IFS), identification of RET gene alteration is important for its unique therapeutic implications.


Subject(s)
Biomarkers, Tumor/genetics , Fibrosarcoma/genetics , Proto-Oncogene Proteins c-ret/genetics , Soft Tissue Neoplasms/genetics , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male , Oncogene Fusion/genetics , Oncogene Proteins, Fusion/genetics
15.
Am J Med Genet A ; 182(4): 673-680, 2020 04.
Article in English | MEDLINE | ID: mdl-31961069

ABSTRACT

Tatton-Brown Rahman syndrome (TBRS) is an overgrowth-intellectual disability syndrome caused by heterozygous variants in DNMT3A. Seventy-eight individuals have been reported with a consistent phenotype of somatic overgrowth, mild to moderate intellectual disability, and similar dysmorphisms. We present six individuals with TBRS, including the youngest individual thus far reported, first individual to be diagnosed with tumor testing and two individuals with variants at the Arg882 domain, bringing the total number of reported cases to 82. Patients reported herein have additional clinical features not previously reported in TBRS. One patient had congenital diaphragmatic hernia. One patient carrying the recurrent p.Arg882His DNMT3A variant, who was previously reported as having a phenotype due to a truncating variant in the CLTC gene, developed a ganglioneuroblastoma at 18 months and T-cell lymphoblastic lymphoma at 6 years of age. Four patients manifested symptoms suggestive of autonomic dysfunction, including central sleep apnea, postural orthostatic hypotension, and episodic vasomotor instability in the extremities. We discuss the molecular and clinical findings in our patients with TBRS in context of existing literature.


Subject(s)
Abnormalities, Multiple/pathology , DNA (Cytosine-5-)-Methyltransferases/genetics , Growth Disorders/pathology , Intellectual Disability/pathology , Mutation , Abnormalities, Multiple/genetics , Adolescent , Adult , Child , Child, Preschool , Clathrin Heavy Chains/genetics , DNA Methyltransferase 3A , Female , Growth Disorders/genetics , Humans , Infant , Intellectual Disability/genetics , Male , Phenotype , Syndrome , Young Adult
16.
Pediatr Blood Cancer ; 67(6): e28276, 2020 06.
Article in English | MEDLINE | ID: mdl-32196952

ABSTRACT

BACKGROUND: Genetic alterations in multiple cell signaling pathways are involved in the molecular pathogenesis of thyroid cancer. Oncogene mutation testing and gene-expression profiling are routinely used for the preoperative risk management of adult thyroid nodules. In this study, we evaluated the potential value of miRNA biomarkers for the classification of pediatric thyroid lesions. PROCEDURE: Double-blind case-control study with 113 resected pediatric lesions: 66 malignant and 47 benign. Quantitative and qualitative molecular data generated with a 10-miRNA expression panel (ThyraMIR) and a next-generation sequencing oncogene panel (ThyGeNEXT) were compared with clinicopathological parameters. RESULTS: miRNAs were differentially expressed in benign versus malignant tumors with distinct expression patterns in different histopathology categories. The 10-miRNA classifier identified 39 (59%) malignant lesions with 100% specificity. A positive classifier score was associated with lymph node metastasis, extrathyroidal extension and intrathyroidal spread. Genetic alterations associated with increased risk for malignancy were detected in 35 (53%) malignant cases, 20 positive for point mutations in BRAF, HRAS, KRAS, NRAS, PIK3CA, or TERT and 15 positive for gene rearrangements involving ALK, NTRK3, PPARG, or RET. The 10-miRNA classifier correctly identified 11 mutation-negative malignant cases. The performance of the combined molecular test was 70% sensitivity and 96% specificity with an area under the curve of 0.924. CONCLUSIONS: These data suggest that the regulatory miRNA pathways underlying thyroid tumorigenesis are similar in adults and children. miRNA expression can identify malignant lesions with high specificity, augment the diagnostic yield of mutation testing, and improve the molecular classification of pediatric thyroid nodules.


Subject(s)
Biomarkers, Tumor/genetics , DNA Mutational Analysis/methods , MicroRNAs/genetics , Mutation , Thyroid Neoplasms/classification , Thyroid Neoplasms/diagnosis , Adolescent , Case-Control Studies , Child , Child, Preschool , Double-Blind Method , Female , Follow-Up Studies , High-Throughput Nucleotide Sequencing , Humans , Male , Prognosis , Thyroid Neoplasms/genetics
17.
Hum Genet ; 138(11-12): 1301-1311, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31686214

ABSTRACT

Haploinsufficiency of FOXF1 causes alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV), a lethal neonatal lung developmental disorder. We describe two similar heterozygous CNV deletions involving the FOXF1 enhancer and re-analyze FOXF1 missense mutation, all associated with an unexpectedly mitigated disease phenotype. In one case, the deletion of the maternal allele of the FOXF1 enhancer caused pulmonary hypertension and histopathologically diagnosed MPV without the typical ACD features. In the second case, the deletion of the paternal enhancer resulted in ACDMPV rather than the expected neonatal lethality. In both cases, FOXF1 expression in lung tissue was higher than usually seen or expected in patients with similar deletions, suggesting an increased activity of the remaining allele of the enhancer. Sequencing of these alleles revealed two rare SNVs, rs150502618-A and rs79301423-T, mapping to the partially overlapping binding sites for TFAP2s and CTCF in the core region of the enhancer. Moreover, in a family with three histopathologically-diagnosed ACDMPV siblings whose missense FOXF1 mutation was inherited from the healthy non-mosaic carrier mother, we have identified a rare SNV rs28571077-A within 2-kb of the above-mentioned non-coding SNVs in the FOXF1 enhancer in the mother, that was absent in the affected newborns and 13 unrelated ACDMPV patients with CNV deletions of this genomic region. Based on the low population frequencies of these three variants, their absence in ACDMPV patients, the results of reporter assay, RNAi and EMSA experiments, and in silico predictions, we propose that the described SNVs might have acted on FOXF1 enhancer as hypermorphs.


Subject(s)
Enhancer Elements, Genetic , Forkhead Transcription Factors/genetics , Mutation, Missense , Persistent Fetal Circulation Syndrome/prevention & control , Polymorphism, Single Nucleotide , Sequence Deletion , Adult , Child , Female , Genomic Imprinting , Humans , Infant, Newborn , Persistent Fetal Circulation Syndrome/genetics , Persistent Fetal Circulation Syndrome/pathology , Phenotype , Prognosis
18.
Pediatr Blood Cancer ; 65(10): e27296, 2018 10.
Article in English | MEDLINE | ID: mdl-29932284

ABSTRACT

Beckwith-Wiedemann syndrome (BWS) is a genetic syndrome associated with overgrowth and cancer predisposition, including predisposition to Wilms tumor (WT). Patients with BWS and BWS spectrum are screened from birth to age 7 years for BWS-associated cancers. However, in some cases a BWS-associated cancer may be the first recognized manifestation of the syndrome. We describe 12 patients diagnosed with BWS after presenting with a WT. We discuss the features of BWS in these patients and hypothesize that earlier detection of BWS by attention to its subtler manifestations could lead to earlier detection of children at risk for associated malignancies.


Subject(s)
Beckwith-Wiedemann Syndrome/complications , Beckwith-Wiedemann Syndrome/diagnosis , Kidney Neoplasms/genetics , Wilms Tumor/genetics , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male
19.
Ann Hepatol ; 16(3): 465-468, 2017.
Article in English | MEDLINE | ID: mdl-28425419

ABSTRACT

Progressive Familial Intrahepatic Cholestasis type 2 (PFIC2) is a rare cholestatic disorder diagnosed in infancy or childhood that can lead to severe hepatic fibrosis and liver failure. Mutations in the ABCB11 gene result in a deficiency of the bile salt export protein (BSEP) and accumulation of bile inside the hepatocytes. Hepatocellular carcinoma is another condition associated with severe forms of deletion mutations in the ABCB11 gene. Treatment options including ursodeoxycholic acid biliary diversion have mixed outcomes and some patients require liver transplantation. Here, we describe two siblings with an extremely mild form of PFIC2 inherited from heterozygous parents. The elder sibling had acute liver failure at the age of six months and both siblings had pruritus, cholestasis, coagulopathy and fat-soluble-vitamin deficiencies in infancy but have been asymptomatic past infancy. Genetic testing of the siblings revealed that each were compound heterozygotes for two missense mutations of the ABCB11 gene: p.C68Y and p.R832H. Medical treatment typical for PFIC2 has not been necessary for either patient. This is the first report of these variants following a mild course in two affected patients.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Cholestasis, Intrahepatic/genetics , Mutation, Missense , ATP Binding Cassette Transporter, Subfamily B, Member 11 , Biopsy, Large-Core Needle , Child , Child, Preschool , Cholestasis, Intrahepatic/diagnosis , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Heredity , Heterozygote , Humans , Immunohistochemistry , Male , Microscopy, Electron, Transmission , Pedigree , Phenotype , Severity of Illness Index
20.
Cytogenet Genome Res ; 150(3-4): 162-175, 2016.
Article in English | MEDLINE | ID: mdl-28002823

ABSTRACT

Genomic alterations are important biological markers for cancer diagnosis and prognosis, disease classification, risk stratification, and treatment selection. Chromosomal microarray analysis (CMA) and next-generation sequencing (NGS) technologies are superb new tools for evaluating cancer genomes. These state-of-the-art technologies offer high-throughput, highly accurate, targeted and whole-genome evaluation of genomic alterations in tumor tissues. The application of CMA and NGS technologies in cancer research has generated a wealth of useful information about the landscape of genomic alterations in cancer and their implications in cancer care. As the knowledge base in cancer genomics and genome biology grows, the focus of research is now shifting toward the clinical applications of these technologies to improve patient care. Although not yet standard of care in cancer, there is an increasing interest among the cancer genomics communities in applying these new technologies to cancer diagnosis in the Clinical Laboratory Improvement Amendments (CLIA)-certified laboratories. Many clinical laboratories have already started adopting these technologies for cancer genomic analysis. We anticipate that CMA and NGS will soon become the major diagnostic means for cancer genomic analysis to meet the increasing demands of precision cancer care.


Subject(s)
Genomics , Neoplasms/therapy , Chromosome Mapping , High-Throughput Nucleotide Sequencing , Humans , Neoplasms/genetics , Precision Medicine
SELECTION OF CITATIONS
SEARCH DETAIL