Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 139(2): 256-280, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34727172

ABSTRACT

ALK-positive histiocytosis is a rare subtype of histiocytic neoplasm first described in 2008 in 3 infants with multisystemic disease involving the liver and hematopoietic system. This entity has subsequently been documented in case reports and series to occupy a wider clinicopathologic spectrum with recurrent KIF5B-ALK fusions. The full clinicopathologic and molecular spectra of ALK-positive histiocytosis remain, however, poorly characterized. Here, we describe the largest study of ALK-positive histiocytosis to date, with detailed clinicopathologic data of 39 cases, including 37 cases with confirmed ALK rearrangements. The clinical spectrum comprised distinct clinical phenotypic groups: infants with multisystemic disease with liver and hematopoietic involvement, as originally described (Group 1A: 6/39), other patients with multisystemic disease (Group 1B: 10/39), and patients with single-system disease (Group 2: 23/39). Nineteen patients of the entire cohort (49%) had neurologic involvement (7 and 12 from Groups 1B and 2, respectively). Histology included classic xanthogranuloma features in almost one-third of cases, whereas the majority displayed a more densely cellular, monomorphic appearance without lipidized histiocytes but sometimes more spindled or epithelioid morphology. Neoplastic histiocytes were positive for macrophage markers and often conferred strong expression of phosphorylated extracellular signal-regulated kinase, confirming MAPK pathway activation. KIF5B-ALK fusions were detected in 27 patients, whereas CLTC-ALK, TPM3-ALK, TFG-ALK, EML4-ALK, and DCTN1-ALK fusions were identified in single cases. Robust and durable responses were observed in 11/11 patients treated with ALK inhibition, 10 with neurologic involvement. This study presents the existing clinicopathologic and molecular landscape of ALK-positive histiocytosis and provides guidance for the clinical management of this emerging histiocytic entity.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/analysis , Histiocytic Disorders, Malignant/drug therapy , Histiocytic Disorders, Malignant/pathology , Protein Kinase Inhibitors/therapeutic use , Adolescent , Adult , Anaplastic Lymphoma Kinase/genetics , Child , Child, Preschool , Female , Histiocytic Disorders, Malignant/complications , Histiocytic Disorders, Malignant/genetics , Humans , Infant , Male , Nervous System Diseases/etiology , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Oncogene Proteins, Fusion/analysis , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Retrospective Studies , Young Adult
3.
Blood ; 137(4): 485-492, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33067622

ABSTRACT

Erdheim-Chester disease (ECD) is a clonal hematopoietic disorder characterized by the accumulation of foamy histiocytes within organs (in particular, frequent retroperitoneal involvement) and a high frequency of BRAFV600E mutations. Although ECD is not commonly recognized to have overt peripheral blood (PB) or bone marrow (BM) disease, we recently identified that ECD patients have a high frequency of a concomitant myeloid malignancy. We thus conducted a systematic clinical and molecular analysis of the BM from 120 ECD patients. Surprisingly, 42.5% of ECD patients (51 of 120) had clonal hematopoiesis whereas 15.8% of patients (19 of 120) developed an overt hematologic malignancy (nearly all of which were a myeloid neoplasm). The most frequently mutated genes in BM were TET2, ASXL1, DNMT3A, and NRAS. ECD patients with clonal hematopoiesis were more likely to be older (P < .0001), have retroperitoneal involvement (P = .02), and harbor a BRAFV600E mutation (P = .049) than those without clonal hematopoiesis. The presence of the TET2 mutation was associated with a BRAFV600E mutation in tissue ECD lesions (P = .0006) and TET2-mutant ECD patients were more likely to have vascular involvement than TET2 wild-type ECD patients. Clonal hematopoiesis mutations in ECD were detected in cells derived from CD34+CD38- BM progenitors and PB monocytes but less frequently present in PB B and T lymphocytes. These data identify a heretofore unrecognized high frequency of clonal hematopoiesis in ECD patients, reaffirm the development of additional high risk of myeloid neoplasms in ECD, and provide evidence of a BM-based precursor cell of origin for many patients with ECD.


Subject(s)
Clonal Hematopoiesis , Erdheim-Chester Disease/physiopathology , Abnormal Karyotype , Adult , Age Factors , Aged , Bone Marrow/pathology , Cell Transformation, Neoplastic/genetics , Clonal Hematopoiesis/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Disease Progression , Erdheim-Chester Disease/genetics , Exons/genetics , Female , Genes, Neoplasm , Humans , Leukemia, Myeloid/genetics , Male , Middle Aged , Multiple Myeloma/genetics , Mutation , Myelodysplastic Syndromes/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Organ Specificity , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics
4.
Am J Hematol ; 98(7): 1058-1069, 2023 07.
Article in English | MEDLINE | ID: mdl-37115038

ABSTRACT

The spectrum of somatic mutations in pediatric histiocytoses and their clinical implications are not fully characterized, especially for non-Langerhans cell histiocytosis (-LCH) subtypes. A cohort of 415 children with histiocytosis from the French histiocytosis registry was reviewed and analyzed for BRAFV600E . Most BRAFWT samples were analyzed by next-generation sequencing (NGS) with a custom panel of genes for histiocytosis and myeloid neoplasia. Of 415 case samples, there were 366 LCH, 1 Erdheim-Chester disease, 21 Rosai-Dorfman disease (RDD), 21 juvenile xanthogranuloma (JXG, mostly with severe presentation), and 6 malignant histiocytosis (MH). BRAFV600E was the most common mutation found in LCH (50.3%, n = 184). Among 105 non-BRAFV600E -mutated LCH case samples, NGS revealed mutations as follows: MAP2K1 (n = 44), BRAF exon 12 deletions (n = 26), and duplications (n = 8), other BRAF V600 codon mutation (n = 4), and non-MAP-kinase pathway genes (n = 5). Wild-type sequences were identified in 17.1% of samples. BRAFV600E was the only variant significantly correlated with critical presentations: organ-risk involvement and neurodegeneration. MAP-kinase pathway mutations were identified in seven RDD (mostly MAP2K1) and three JXG samples, but most samples were wild-type on NGS. Finally, two MH samples had KRAS mutations, and one had a novel BRAFG469R mutation. Rarely, we identified mutations unrelated to MAP-kinase pathway genes. In conclusion, we characterized the mutational spectrum of childhood LCH and clinical correlations of variants and subtypes. Variants responsible for JXG and RDD were not elucidated in more than half of the cases, calling for other sequencing approaches.


Subject(s)
Erdheim-Chester Disease , Histiocytosis, Langerhans-Cell , Humans , Child , Histiocytosis, Langerhans-Cell/genetics , Proto-Oncogene Proteins B-raf/genetics , Erdheim-Chester Disease/genetics , Mutation , Exons
5.
Genes Chromosomes Cancer ; 60(11): 723-732, 2021 11.
Article in English | MEDLINE | ID: mdl-34245196

ABSTRACT

Glomus tumors (GTs) are perivascular tumors mostly occurring in the distal extremities. Rare cases arise in the digestive tract and may be misdiagnosed with neuroendocrine or gastrointestinal stromal tumors. We aimed to specify the features of GT of the upper digestive tract. Clinical, histological, phenotypic, and molecular features of 16 digestive GTs were analyzed, of whom two underwent whole exome and RNA sequencing to search for gene alterations. RNA-sequencing disclosed a t(1:5)(p13;q32) translocation, which resulted in the fusion of CARMN and NOTCH2 in two GTs. The fusion gene encoded a protein sequence corresponding to the NOTCH2 intracellular domain that functions as transcription factor. These finding was supported by high expression of genes targeted by NOTCH. The CARMN-NOTCH2 translocation was detected in 14 out of 16 (88%) GTs of the upper digestive tract; but in only in two out of six cutaneous GTs (33%). Most digestive GT arose from the stomach (n = 13), and the others from duodenal (2) or oesophagous (1). Nuclear expression of NOTCH2 was detected in the 14 cases containing the fusion transcripts. The CARMN-NOTCH2 fusion transcript may contribute to activation of the NOTCH2 pathway in GT and drive tumor development. The high frequency of this translocation in GT of the upper digestive track suggest that detection of nuclear NOTCH2 expression may be useful diagnostic biomarker of these tumors.


Subject(s)
Biomarkers, Tumor/genetics , Gastrointestinal Neoplasms/genetics , Gene Fusion , Glomus Tumor/genetics , MicroRNAs/genetics , Receptor, Notch2/genetics , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Glomus Tumor/metabolism , Glomus Tumor/pathology , Humans
6.
Br J Haematol ; 194(4): 745-749, 2021 08.
Article in English | MEDLINE | ID: mdl-34312844

ABSTRACT

BRAF inhibitors are an effective treatment for BRAFV600E -mutated, risk-organ-positive Langerhans cell histiocytosis (RO+ LCH). However, cell-free BRAFV600E DNA often persists during therapy and recurrence frequently occurs after therapy discontinuation. To identify a pathological reservoir of BRAFV600E -mutated cells, we studied peripheral blood cells obtained from six infants with RO+ multisystem (MS) LCH that received targeted therapy. After cell sorting, the BRAFV600E mutation was detected in monocytes (n = 5), B lymphocytes (n = 3), T lymphocytes (n = 2), and myeloid and plasmacytoid dendritic cells (n = 2 each). This biomarker may offer an interesting tool for monitoring the effectiveness of new therapeutic approaches for weaning children with RO+ LCH from targeted therapy.


Subject(s)
Histiocytosis, Langerhans-Cell/drug therapy , Point Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Child , Child, Preschool , Histiocytosis, Langerhans-Cell/blood , Histiocytosis, Langerhans-Cell/genetics , Humans , Infant , Point Mutation/drug effects , Proto-Oncogene Proteins B-raf/blood
7.
Blood ; 130(8): 1007-1013, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28679734

ABSTRACT

Erdheim-Chester disease (ECD) is a rare non-Langerhans cell histiocytosis that most commonly affects adults and is driven by a high frequency of mutations in BRAF, MAP2K1, and kinases promoting MAPK signaling. Because of the relative rarity of ECD, key clinical features of the disease may not be well defined. Across a multi-institutional cohort of 189 patients with ECD and ECD overlapping with Langerhans cell histiocytosis (so-called mixed histiocytosis [MH]), we identified an unexpected and heretofore undescribed frequent occurrence of myeloid neoplasms among patients with ECD and MH. Some 10.1% (19/189) of patients with ECD have an overlapping myeloid neoplasm, most commonly occurring as a myeloproliferative neoplasm (MPN), myelodysplastic syndrome (MDS), or mixed MDS/MPN overlap syndrome (including chronic myelomonocytic leukemia). Consistent with this, molecular analysis frequently detected hallmark driver mutations of myeloid neoplasms (such as JAK2V617F and CALR mutations) coexisting with those characteristic of histiocytosis (such as BRAFV600E and MAP2K1 mutations). Histiocytosis patients diagnosed with a concomitant myeloid malignancy were significantly older at diagnosis and more commonly presented with MH than those without a myeloid malignancy. In some cases, the presence of distinct kinase mutations in the histiocytosis and myeloid neoplasm resulted in discordant and adverse responses to kinase-directed targeted therapies. These data highlight the clinical importance of evaluating adults with histiocytosis for a concomitant myeloid neoplasm.


Subject(s)
Bone Marrow Neoplasms/complications , Bone Marrow Neoplasms/epidemiology , Histiocytosis, Non-Langerhans-Cell/complications , Adult , Aged , Erdheim-Chester Disease/complications , Female , Humans , Male , Middle Aged , Molecular Targeted Therapy , Prevalence
8.
Blood ; 130(2): 176-180, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28566492

ABSTRACT

Langerhans cell histiocytosis (LCH) and the non-LCH neoplasm Erdheim-Chester disease (ECD) are heterogeneous neoplastic disorders marked by infiltration of pathologic macrophage-, dendritic cell-, or monocyte-derived cells in tissues driven by recurrent mutations activating MAPK signaling. Although recent data indicate that at least a proportion of LCH and ECD patients have detectable activating kinase mutations in circulating hematopoietic cells and bone marrow-based hematopoietic progenitors, functional evidence of the cell of origin of histiocytosis from actual patient materials has long been elusive. Here, we provide evidence for mutations in MAPK signaling intermediates in CD34+ cells from patients with ECD and LCH/ECD, including detection of shared origin of LCH and acute myelomonocytic leukemia driven by TET2-mutant CD34+ cell progenitors in one patient. We also demonstrate functional self-renewal capacity for CD34+ cells to drive the development of histiocytosis in xenotransplantation assays in vivo. These data indicate that the cell of origin of at least a proportion of patients with systemic histiocytoses resides in hematopoietic progenitor cells prior to committed monocyte/macrophage or dendritic cell differentiation and provide the first example of a patient-derived xenotransplantation model for a human histiocytic neoplasm.


Subject(s)
Bone Marrow Cells/pathology , DNA-Binding Proteins/genetics , Erdheim-Chester Disease/pathology , Hematopoietic Stem Cells/pathology , Histiocytosis, Langerhans-Cell/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins/genetics , Adult , Alleles , Animals , Antigens, CD34/genetics , Antigens, CD34/immunology , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Cell Differentiation , DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dioxygenases , Erdheim-Chester Disease/genetics , Erdheim-Chester Disease/immunology , Gene Expression , Hematopoietic Stem Cells/immunology , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/immunology , Humans , Immunophenotyping , Macrophages/immunology , Macrophages/pathology , Mice , Monocytes/immunology , Monocytes/pathology , Mutation , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins B-raf/immunology , Transplantation, Heterologous
9.
Am J Dermatopathol ; 41(1): 29-36, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30252693

ABSTRACT

Langerhans cell (LC) histiocytoma is a neonatal tumor that often consists of a single, ulcerated nodule. Systemic involvement is rare, and LC histiocytoma is considered to be a variant of congenital, self-healing LC histiocytosis (also referred to as Hashimoto-Pritzker disease). In view of its low prevalence, LC histiocytoma is not always diagnosed in a clinical examination and requires histological confirmation. Furthermore, the histological and molecular features of LC histiocytoma have not been well characterized. Here, we report on 6 cases of this rare disease and review the corresponding literature. LC histiocytoma differs from classical self-healing LC histiocytosis with regard to the pathological features; we found that LC histiocytoma was associated with massive infiltration by histiocytes of various sizes and shapes (although often large) throughout the dermis and the superficial subcutis. Epidermotropism was rare, mitotic figures were not inconspicuous, and necrotic or calcified areas were often present. Immunohistochemical assessment revealed a mixture of different types of histiocytes (with CD1a CD207, CD1a CD207, and CD1a CD207 CD163 cells). Genetic testing was performed in 5 cases; it revealed a BRAF mutation (p.V600E and p.485_490delinsF) in 2 cases, a HRAS mutation (p.T58I) in 1 case, a combination of 2 PTEN mutations in another case (p.I224M and p. R234W), and no mutations in the fifth case. All the lesions regressed spontaneously, and none recurred during follow-up.


Subject(s)
Histiocytes/pathology , Histiocytoma, Benign Fibrous/pathology , Histiocytosis, Langerhans-Cell/pathology , Langerhans Cells/pathology , Skin Neoplasms/pathology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Female , Genetic Predisposition to Disease , Histiocytes/chemistry , Histiocytoma, Benign Fibrous/chemistry , Histiocytoma, Benign Fibrous/genetics , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/metabolism , Humans , Infant , Infant, Newborn , Langerhans Cells/chemistry , Male , Mutation , Neoplasm Regression, Spontaneous , Phenotype , Skin Neoplasms/chemistry , Skin Neoplasms/genetics
10.
Br J Haematol ; 183(4): 608-617, 2018 11.
Article in English | MEDLINE | ID: mdl-30421536

ABSTRACT

Neurodegenerative (ND) complications in Langerhans cell histiocytosis (LCH) are a late-onset but dramatic sequelae for which incidence and risk factors are not well defined. Based on a national prospective registry of paediatric LCH patients, we determined the incidence rate of clinical ND LCH (cND-LCH) and analysed risk factors, taking into account disease extent and molecular characteristics. Among 1897 LCH patients, 36 (1·9%) were diagnosed with a cND-LCH. The 10-year cumulative incidence of cND-LCH was 4·1%. cND-LCH typically affected patients previously treated for a multisystem, risk organ-negative LCH, represented in 69·4% of cND-LCH cases. Pituitary gland, skin and base skull/orbit bone lesions were more frequent (P < 0·001) in cND-LCH patients compared to those without cND-LCH (respectively 86·1% vs. 12·2%, 75·0% vs. 34·2%, and 63·9% vs. 28·4%). The 'cND susceptible patients' (n = 671) i.e., children who had experienced LCH disease with pituitary or skull base or orbit bone involvement, had a 10-year cND risk of 7·8% vs. 0% for patients who did not meet these criteria. Finally, BRAFV600E status added important information among these cND susceptible patients, with the 10-year cND risk of 33·1% if a BRAFV600E mutation was present compared to 2·9% if it was absent (P = 0·002).


Subject(s)
Histiocytosis, Langerhans-Cell/epidemiology , Neurodegenerative Diseases/epidemiology , Registries , Adolescent , Child , Child, Preschool , Female , Follow-Up Studies , Histiocytosis, Langerhans-Cell/metabolism , Histiocytosis, Langerhans-Cell/pathology , Humans , Incidence , Infant , Infant, Newborn , Longitudinal Studies , Male , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Risk Factors
11.
Cancer Invest ; 36(6): 338-348, 2018.
Article in English | MEDLINE | ID: mdl-30136875

ABSTRACT

Aneuploidy is a common feature of cancer cells and may contribute to cellular transformation and cancer development. In this study, we found that significant down-regulation of CDKN2A, CHEK2, CDCA8, TP53BP1, and CCNDBP1 led to chromosome imbalances in two diploid non-immortalized human cell lines; however, only CDKN2A inhibition enhanced cell proliferation and additionally up-regulated three cell cycle control genes: CDCA8, AURKA, and CCND. These results confirm that CDKN2A is a tumor suppressor gene driving human cancer development by inducing cell aneuploidy and cell cycle up-regulation.


Subject(s)
Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p18/genetics , Genes, Tumor Suppressor , Aneuploidy , Aurora Kinase A/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Checkpoint Kinase 2/genetics , Cyclin-Dependent Kinase Inhibitor p16 , Gene Expression Regulation, Neoplastic , Humans , Tumor Suppressor p53-Binding Protein 1/genetics
12.
J Pathol ; 242(1): 10-15, 2017 05.
Article in English | MEDLINE | ID: mdl-28127763

ABSTRACT

Germline alterations in DNA repair genes are implicated in cancer predisposition and can result in characteristic mutational signatures. However, specific mutational signatures associated with base excision repair (BER) defects remain to be characterized. Here, by analysing a series of colorectal cancers (CRCs) using exome sequencing, we identified a particular spectrum of somatic mutations characterized by an enrichment of C > A transversions in NpCpA or NpCpT contexts in three tumours from a MUTYH-associated polyposis (MAP) patient and in two cases harbouring pathogenic germline MUTYH mutations. In two series of adrenocortical carcinomas (ACCs), we identified four tumours with a similar signature also presenting germline MUTYH mutations. Taken together, these findings demonstrate that MUTYH inactivation results in a particular mutational signature, which may serve as a useful marker of BER-related genomic instability in new cancer types. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/genetics , Colorectal Neoplasms/genetics , DNA Glycosylases/genetics , Mutation , Animals , DNA Glycosylases/deficiency , DNA Mutational Analysis/methods , DNA, Neoplasm/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Mice, Knockout , Transcriptome/genetics
13.
Mol Cancer ; 16(1): 115, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28679432

ABSTRACT

Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasia with constitutive activation of the MAPKinase RAS-RAF-MEK-ERK cell signaling pathway. We analyzed 9 LCH cases without BRAF V600 and MAP2K1 mutations by whole exome sequencing. We identified a new somatic BRAF splicing mutation in 2 cases. Both cases were childhood single system (SS) LCH cases, with self-healing outcome of the bone lesions. This mutant consisted in a 9 base pair duplication (c.1511_1517 + 2 duplication), encoding for a predicted mutant protein with insertion of 3 amino acids (p.Arg506_Lys507insLeuLeuArg) in the N-terminal lobe of the kinase domain of BRAF. Transient expression of the c.1511_1517 + 2dup BRAF mutant in HEK293 cells enhanced MAPKinase pathway activation, and was not inhibited by vemurafenib but was inhibited by PLX8394, a second-generation BRAF inhibitor able to inhibit signaling of BRAF monomers and dimers. Future LCH molecular screening panel should include this new mutation to better define its prevalence in LCH and its restriction to autoregressive bone SS LCH.


Subject(s)
Histiocytosis, Langerhans-Cell/genetics , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , RNA Splicing/genetics , Adolescent , Base Sequence , Child , Child, Preschool , Exons/genetics , Female , Gene Duplication , Histiocytosis, Langerhans-Cell/drug therapy , Humans , Infant , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase Inhibitors/therapeutic use
14.
Br J Haematol ; 178(3): 457-467, 2017 08.
Article in English | MEDLINE | ID: mdl-28444728

ABSTRACT

The BRAFV600E mutation is reported in half of patients with Langerhans cell histiocytosis (LCH). This study investigated the detection of the BRAFV600E allele in circulating cell-free (ccf) DNA in a paediatric LCH cohort. Children with BRAFV600E -mutated LCH were investigated to detect ccf BRAFV600E at diagnosis (n = 48) and during follow-up (n = 17) using a picolitre-droplet digital PCR assay. At diagnosis, ccf BRAFV600E was positive in 15/15 (100%) patients with risk-organ positive multisystem (RO+ MS) LCH, 5/12 (42%) of patients with RO- MS LCH and 3/21 (14%) patients with single-system (SS) LCH (P < 0·001, Fisher's exact test). The positive BRAFV600E load was higher for RO+ patients (mean, 2·90%; range, 0·04-11·4%) than for RO- patients (mean, 0·16%; range, 0·01-0·39) (P = 0·003, Mann-Whitney U test). After first-line vinblastine-steroid induction therapy, 7/7 (100%) of the non-responders remained positive for ccf BRAFV600E compared to 2/4 (50%) of the partial-responders and 0/4 of the complete responders (P = 0·002, Fisher's exact test). Six children treated with vemurafenib showed a clinical response that was associated with a decrease in the ccf BRAFV600E load at day 15. Thus, ccf BRAFV600E is a promising biomarker for monitoring the response to therapy for children with RO+ MS LCH or RO- LCH resistant to first-line chemotherapy.


Subject(s)
Histiocytosis, Langerhans-Cell/diagnosis , Proto-Oncogene Proteins B-raf/blood , Adolescent , Alleles , Biomarkers/blood , Cell-Free System/metabolism , Child , Child, Preschool , Drug Monitoring/methods , Drug Therapy, Combination , Female , Follow-Up Studies , Glucocorticoids/therapeutic use , Histiocytosis, Langerhans-Cell/drug therapy , Histiocytosis, Langerhans-Cell/genetics , Humans , Indoles/therapeutic use , Infant , Male , Mutation , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/therapeutic use , Vemurafenib , Vinblastine/therapeutic use
15.
BMC Dermatol ; 17(1): 9, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28668077

ABSTRACT

BACKGROUND: Somatic mutations of BRAF or NRAS activating the MAP kinase cell signaling pathway are present in 70% of cutaneous melanomas. The mutant allele frequency of BRAF V600E (M%BRAF) was recently shown to be highly heterogeneous in melanomas. The present study focuses on the NRAS Q61 mutant allele frequency (M%NRAS). METHODS: Retrospective quantitative analyze of 104 NRAS mutated melanomas was performed using pyrosequencing. Mechanisms of M%NRAS imbalance were studied by fluorescence in situ hybridization (FISH) and microsatellite analysis. RESULTS: M%NRAS was increased in 27.9% of cases. FISH revealed that chromosome 1 instability was the predominant mechanism of M%NRAS increase, with chromosome 1 polysomy observed in 28.6% of cases and intra-tumor cellular heterogeneity with copy number variations of chromosome 1/NRAS in 23.8%. Acquired copy-neutral loss of heterozygosity (LOH) was less frequent (19%). However, most samples with high M%NRAS had only one copy of NRAS locus surrounding regions suggesting a WT allele loss. Clinical characteristics and survival of patients with either <60% or ≥60% of M%NRAS were not different. CONCLUSION: As recently shown for M%BRAF, M%NRAS is highly heterogeneous. The clinical impacts of high M%NRAS should be investigated in a larger series of patients.


Subject(s)
Chromosomes, Human, Pair 1 , GTP Phosphohydrolases/genetics , Gene Frequency , Melanoma/genetics , Membrane Proteins/genetics , Mutation , Skin Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Chromosome Aberrations , DNA Copy Number Variations , Female , Humans , Kaplan-Meier Estimate , Male , Melanoma/mortality , Middle Aged , Retrospective Studies , Skin Neoplasms/mortality , Melanoma, Cutaneous Malignant
16.
Blood ; 124(19): 3016-9, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25150293

ABSTRACT

Erdheim-Chester disease (ECD) is a rare histiocytic disorder that is challenging to diagnose and treat. We performed molecular analysis of BRAF in the largest cohort of ECD patients studied to date followed by N/KRAS, PIK3CA, and AKT1 mutational analysis in BRAF wild-type patients. Forty-six of 80 (57.5%) of patients were BRAFV600E-mutant. NRAS mutations were detected in 3 of 17 ECD BRAFV600E wild-type patients. PIK3CA mutations (p.E542K, p.E545K, p.A1046T, and p.H1047R) were detected in 7 of 55 patients, 4 of whom also had BRAF mutations. Mutant NRAS was present in peripheral blood CD14(+) cells, but not lymphoid cells, from an NRASQ61R mutant patient. Our results underscore the central role of RAS-RAF-MEK-ERK activation in ECD and identify an important role of activation of RAS-PI3K-AKT signaling in ECD. These results provide a rationale for targeting mutant RAS or PI3K/AKT/mTOR signaling in the subset of ECD patients with NRAS or PIK3CA mutations.


Subject(s)
Erdheim-Chester Disease/genetics , GTP Phosphohydrolases/genetics , Membrane Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Adult , Aged , Aged, 80 and over , Class I Phosphatidylinositol 3-Kinases , Erdheim-Chester Disease/metabolism , Female , GTP Phosphohydrolases/metabolism , Histiocytes/metabolism , Humans , MAP Kinase Signaling System/genetics , Male , Membrane Proteins/metabolism , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Point Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras) , Recurrence , ras Proteins/genetics , ras Proteins/metabolism
17.
Exp Dermatol ; 25(6): 472-4, 2016 06.
Article in English | MEDLINE | ID: mdl-26990546

ABSTRACT

One-fifth of cutaneous melanomas have dominant gain-of-function mutations of the NRAS oncogene. We report the first two cases of increasing NRAS mutant allele frequency in melanoma metastases and show that the chromosomal mechanism of this homozygosity is an increased polysomy of chromosome 1. We observed an increase in NRAS mutant allele percentage (NRAS-MA%) in the metastatic melanoma progression from 2 patients with melanomas harbouring a NRAS mutation (p.Q61K in case 1 and p.Q61R in case 2). In case 1, we observed a NRAS-MA% increase from 18% within the first metastatic node to 81%, 92% and 85% respectively in the three subsequent metastases: lymph node, brain and subcutaneous metastases biopsied 1, 6 and 17 months, respectively, after the initial lymph node biopsy. In case 2, we observed an increase in NRAS-MA% from 40% within the primary melanoma to 63% within the metastatic lymph node. FISH analysis showed the same results in both cases: a frequent polysomy of chromosome 1 in metastasis samples with NRAS mutant allele percentage >60%, while most cells were disomic in the samples with well-balanced heterozygous mutations. The percentage of NRAS mutant allele may increase during metastatic progression and may be associated with chromosomal instability. Further studies are needed to evaluate the prognostic impact of the NRAS homozygous status and/or polyploidy in metastatic cutaneous melanomas.


Subject(s)
Chromosome Duplication , Chromosomes, Human, Pair 1 , GTP Phosphohydrolases/genetics , Gain of Function Mutation , Melanoma/genetics , Membrane Proteins/genetics , Aged , Humans , Male , Middle Aged
19.
BMC Cancer ; 15: 497, 2015 Jul 04.
Article in English | MEDLINE | ID: mdl-26141748

ABSTRACT

BACKGROUND: BRAF mutations are present in 40% of human skin melanomas. Mutated tumors with an increased percentage of BRAF mutant alleles (BRAF-M%) may have a better response to RAF/MEK inhibitors. We evaluated the BRAF-M% in melanomas, and the genetic causes of its variation. METHODS: BRAF-M% was quantified by pyrosequencing, real-time PCR (rtPCR) and/or picoliter-droplet PCR (dPCR). BRAF mutant expression was detected by immunohistochemistry. Chromosomal alterations were analyzed with fluorescence in situ hybridization (FISH), and single nucleotide polymorphism (SNP) arrays. RESULTS: BRAF-M% quantification obtained with pyrosequencing was highly correlated (R = 0.94) with rtPCR, and with dPCR. BRAF-M% quantified from DNA and RNA were also highly correlated (R = 0.98). Among 368 samples with >80% tumor cells, 38.6% had a BRAF (V600E) mutation. Only 66.2% cases were heterozygous (BRAF-M% 30 to 60%). Increased BRAF-M% (>60%) was observed in 19% of cases. FISH showed a polysomy of chromosome 7 in 13.6%, 35.3% and 54.5% of BRAF wild-type, heterozygous and non-heterozygous BRAF-mutated samples, respectively (P < 0.005). Amplification (5.6%) and loss (3.2%) of BRAF locus were rare. By contrast, chromosome 7 was disomic in 27/27 BRAF-mutated nevi. CONCLUSIONS: BRAF-M% is heterogeneous and frequently increased in BRAF-mutant melanomas. Aneuploidy of chromosome 7 is more frequent in BRAF mutant melanomas, specifically in those with high BRAF-M%.


Subject(s)
Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics , Aneuploidy , Chromosomes, Human, Pair 7/genetics , Heterozygote , Humans , In Situ Hybridization, Fluorescence , Melanoma/epidemiology , Polymorphism, Single Nucleotide/genetics , Real-Time Polymerase Chain Reaction
20.
Mol Cancer ; 13: 246, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25373456

ABSTRACT

BACKGROUND: A KIT gain of function mutation is present in 70% of gastrointestinal stromal tumors (GISTs) and the wild-type (WT) allele is deleted in 5 to 15% of these cases. The WT KIT is probably deleted during GIST progression. We aimed to identify the mechanism of WT KIT loss and to determine whether other genes are involved or affected. METHODS: Whole-genome SNP array analyses were performed in 22 GISTs with KIT exon 11 mutations, including 11 with WT loss, to investigate the mechanisms of WT allele deletion. CGH arrays and FISH were performed in some cases. Common genetic events were identified by SNP data analysis. The 9p21.3 locus was studied by multiplex quantification of genomic DNA. RESULTS: Chromosome instability involving the whole chromosome/chromosome arm (whole C/CA) was detected in 21/22 cases. The GISTs segregated in two groups based on their chromosome number: polyGISTs had numerous whole C/CA gains (mean 23, range [9 to 43]/3.11 [1 to 5]), whereas biGISTs had fewer aberrations. Whole C/CA losses were also frequent and found in both groups. There were numerous copy-neutral losses of heterozygosity (cnLOH) of whole C/CA in both polyGIST (7/9) and biGIST (9/13) groups. cnLOH were frequent on 4q, 11p, 11q, 1p, 2q, 3p and 10, and never involved 12p, 12q, 20p, 20q or 19q. Other genetic alterations included segmental chromosome abnormalities, complete bi-allelic deletions (homozygous deletions) and, more rarely, amplifications. Nine of 11 GISTs with homozygous KIT exon 11 mutations had cnLOH of chromosome 4. CONCLUSION: The cnLOH of whole C/CA is a frequent genetic alteration in GISTs and is closely associated with homozygous mutations of KIT and WT allele deletion.


Subject(s)
Chromosome Deletion , Gastrointestinal Stromal Tumors/genetics , Loss of Heterozygosity , Adult , Aged , Aged, 80 and over , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Progression , Female , Gastrointestinal Stromal Tumors/pathology , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Polyploidy
SELECTION OF CITATIONS
SEARCH DETAIL