Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Pers Med ; 12(10)2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36294734

ABSTRACT

BACKGROUND: a specific subset of metastatic triple-negative breast cancers (mTNBC) is characterized by homologous recombination deficiency (HRD), leading to enhanced sensitivity to platinum-based chemotherapy. Apart from mutations in BRCA1/2 genes, the evaluation of other HRD-related alterations has been limited to date. As such, we analyzed data from mTNBC patients enrolled in the ProfiLER-01 study to determine the prevalence of alterations in homologous recombination-related (HRR) genes and their association with platinum sensitivity. METHODS: next-generation sequencing and promoter methylation of BRCA1 and RAD51C were performed on tumors from patients with mTNBC, using a panel of 19 HRR genes. Tumors were separated into three groups based on their molecular status: mutations in BRCA1/2, mutations in other HRR genes (BRCA1/2 excluded) or BRCA1/RAD51C promoter methylation and the absence of molecular alterations in HRR genes (groups A, B and C, respectively). Sensitivity to platinum-based chemotherapy was evaluated through the radiological response. RESULTS: mutations in BRCA1/2 were detected in seven (13.5%) patients, while alterations in other HRR genes or hypermethylation in BRCA1 or RAD51C were reported in 16 (30.7%) patients; furthermore, no alteration was found in the majority of patients (n = 29; 55.8%). Among 27 patients who received platinum-based chemotherapy, the disease control rate was 80%, 55% and 18% (groups A, B and C, respectively; p = 0.049). Regarding group B, patients with disease control exhibited mutations in FANCL, FANCA and the RAD51D genes or RAD51C methylation; Conclusion: mutations in HRR genes and epimutations in RAD51C were associated with disease control through platinum-based chemotherapy. As such, apart from well-characterized alterations in BRCA1/2, a more comprehensive evaluation of HRD should be considered in order to enlarge the selection of patients with mTNBC that could benefit from platinum-based chemotherapy.

2.
ESMO Open ; 5(6): e001082, 2020 11.
Article in English | MEDLINE | ID: mdl-33246932

ABSTRACT

BACKGROUND: Imatinib is the standard first-line therapy in metastatic gastrointestinal stromal tumours (GIST). Investigational multi-kinase inhibitors (MKIs) such as nilotinib, dasatinib or masitinib have been tested as first-line therapies in phase II/III studies. This might theoretically result either in increased survival or in early emergence of resistance to approved MKIs. METHODS: To assess whether using MKIs other than imatinib in first line decreases imatinib efficacy in second line for patients with GIST, a retrospective chart review was performed from 2005 to 2011 in two French tertiary centres of patients with GIST who received investigational MKIs (in phase II/III trials) as first-line treatment, followed by imatinib as second line. RESULTS: Of 46 patients, (55% women, median age 55 years (range 24-81)), 22 (47%) had a KIT exon 11 mutation, 1 a KIT exon 9 mutation (2%), 1 a PDGFRA D842V mutation (2%). Out of 46 patients, 21 (46%) received masitinib, 17 (37%) received dasatinib and 8 (17%) received nilotinib as first-line treatment with a median progression-free survival of 18.0 months (95% CI: 8.5 to 25.5). Median time to imatinib failure was 19.7 months (95% CI: 13.5 to 29.0). Median time to second relapse was 48.7 months (95% CI: 31.2 to 72.0). Median overall survival from time of initial metastasis diagnosis was 5.7 years (95% CI: 4.5 to 7.4). CONCLUSIONS: Patients with GIST who received investigational MKIs as first-line treatment and imatinib as second line had a time to second relapse longer than that observed historically with imatinib in first line, suggesting that using MKIs other than imatinib in first line does not decrease the efficacy of subsequent treatment lines.


Subject(s)
Gastrointestinal Stromal Tumors , Adult , Aged , Aged, 80 and over , Female , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Humans , Imatinib Mesylate/therapeutic use , Male , Middle Aged , Mutation , Neoplasm Recurrence, Local , Receptor, Platelet-Derived Growth Factor alpha/genetics , Retrospective Studies , Young Adult
3.
Am J Surg Pathol ; 44(10): 1398-1405, 2020 10.
Article in English | MEDLINE | ID: mdl-32732488

ABSTRACT

We report a series of 6 melanocytic proliferations harboring both NRAS and IDH1 hotspot mutations. Clinically, there was no specific sex-ratio, ages ranged from 18 to 85 years, and the trunk and limbs were the most affected localizations. In half of the cases, progressive modification of a pre-existing nevus was reported. Morphologically, all tumors were predominantly based in the dermis and the most striking pathologic finding was the presence of a background architecture of congenital-type nevi with a superimposed biphasic pattern formed by dendritic pigmented melanocytes surrounding areas of nevoid melanocytes. This finding was further underscored by HMB45 staining, which was positive in the dendritic cells and negative in the nevoid melanocytes. Four cases displayed increased cellularity and 1 case showed increased dermal mitotic activity. DNA and RNA sequencing revealed NRAS and IDH1 comutations in all 6 cases, with homogenous expression data according to unsupervised clustering analysis. Array-comparative genomic hybridization revealed no copy number alteration for the 2 most cellular and mitogenic cases. All were surgically excised, available follow-up for 2 patients showed no relapse nor metastases. We hypothesize that the IDH1 mutation is a secondary event in a pre-existing NRAS-mutated nevus and could be in part responsible for the emergence of a pigmented dendritic dermal component. So far, such comutations have been reported in one benign melanocytic nevus and several melanomas. This combination could represent a new subgroup of intermediate prognosis (melanocytoma) with a distinctive morphology. Further acquisition of genomic anomalies could progressively lead to malignant transformation.


Subject(s)
GTP Phosphohydrolases/genetics , Isocitrate Dehydrogenase/genetics , Melanocytes/pathology , Membrane Proteins/genetics , Nevus, Pigmented/genetics , Skin Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/genetics , Female , Humans , Male , Middle Aged , Nevus, Pigmented/pathology , Skin Neoplasms/pathology , Young Adult
4.
Bull Cancer ; 107(3): 385-390, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32115180

ABSTRACT

The group of rare malignant ovarian tumors includes the group of germ cell tumors, sex cords stromal ovarian tumors, small cell carcinoma, malignant Brenner tumors, rare epithelial tumors such as mucinous carcinoma, clear cell carcinoma, or low-grade serous carcinoma, as well as ovarian carcinosarcoma. Together they comprise about 10% of all ovarian tumors. Due to their low prevalence and their heterogeneity, data and treatment recommendations are limited. Even though all ovarian tumors are staged according to the FIGO staging of epithelial ovarian tumors, treatment differs especially in germ cell tumors and sex cords stromal ovarian tumors. Non-epithelial ovarian tumors can arise from a variety of ovarian precursor cells such as germ cells, granulosa cells, theca cells, or stromal fibroblasts. As can be expected already due to their divergent precursor lesions, these malignancies are substantially different but united by their rarity. This overview article gives a comprehensive summary on the pathology and clinical presentation, as well as therapy recommendations of a selection of those rare ovarian tumors, based on the latest national guidelines and related important publications.


Subject(s)
Ovarian Neoplasms , Rare Diseases , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Clear Cell/therapy , Adenocarcinoma, Mucinous/pathology , Adenocarcinoma, Mucinous/therapy , Brenner Tumor/pathology , Brenner Tumor/therapy , Carcinoma, Small Cell/pathology , Carcinoma, Small Cell/therapy , Carcinosarcoma/pathology , Carcinosarcoma/therapy , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/therapy , Female , Humans , Neoplasms, Germ Cell and Embryonal/pathology , Neoplasms, Germ Cell and Embryonal/therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Rare Diseases/pathology , Rare Diseases/therapy , Sex Cord-Gonadal Stromal Tumors/pathology , Sex Cord-Gonadal Stromal Tumors/therapy
5.
Virchows Arch ; 476(3): 439-443, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31754815

ABSTRACT

We report a series of 21 compound blue nevi, a rare variant in the vast clinical and morphological spectrum of blue melanocytic proliferations. Clinically, they presented in young adults, with a slight female predominance. One-third were located on the dorsum of the foot. Morphologically, all cases displayed large dendritic melanocytes restricted to the deep layers of the epidermis. The compound component was central and evenly distributed. Melanocytic density ranged from scarce isolated cells to a confluent lentiginous architecture. In 12 of the 21 cases, junctional nests of small, bland, weakly pigmented melanocytes were associated. These nests became confluent in the most cellular cases. In all cases, a dermal component was immediately present underneath, mainly of cellular blue nevus-type. All cases were genetically confirmed to harbor either a GNAQ or GNA11 hotspot mutation. This study expands the morphological spectrum of blue nevi that should not be restricted to a strictly intradermal melanocytic proliferation.


Subject(s)
Nevus, Blue/genetics , Nevus, Blue/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Female , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Male , Melanocytes/pathology , Middle Aged , Mutation , Young Adult
6.
Future Oncol ; 15(18): 2139-2149, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31185741

ABSTRACT

Aim: We performed a clinical audit of the management of patients with EGFR mutations, 1 year after the introduction of EGFR tyrosine kinase inhibitor (EGFR-TKI) in first-line treatment. Methods: Compliance was defined by tumor molecular profiling for stage IIIB and IV non-small-cell lung cancer and first-line treatment as recommended by the French guidelines. Results: Among the 169 EGFR-mutated patients, compliance was 76.4%. The most common noncompliance criterion was chemotherapy given in first-line treatment instead of EGFR-TKI. No dedicated multidisciplinary meeting and type of institutions were independent unfavorable predictors for compliance. Compliance to guidelines was significantly correlated with time-to-first subsequent treatment improvement (2.5 vs 9.1 months; p < 0.0001). Conclusion: Implementation of new standards of care is challenging. Our results reinforce the role of multidisciplinary meetings to provide a better access to innovating therapeutics.


Subject(s)
Guideline Adherence , Lung Neoplasms/epidemiology , Molecular Diagnostic Techniques/standards , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/epidemiology , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/therapy , Clinical Audit , Disease Management , Female , France , Genes, erbB-1 , Geography , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/etiology , Lung Neoplasms/therapy , Male , Middle Aged , Molecular Diagnostic Techniques/methods , Molecular Targeted Therapy , Mutation , Neoplasm Metastasis , Neoplasm Staging , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies , Survival Analysis
8.
Pigment Cell Melanoma Res ; 32(5): 708-713, 2019 09.
Article in English | MEDLINE | ID: mdl-30945443

ABSTRACT

A girl, born with a posterior  lumbosacral giant congenital nevus, developed a central nodule that expanded over a period of 14 months into a 10-cm pedunculated mass. Histological analysis of the mass revealed melanoma of myxoid, small round-cell type with areas of  rhabdomyosarcomatous  transformation confirmed by immunohistochemistry. RNA sequencing identified an in-frame SASS6(e14)-RAF1(e8) fusion in both components and the nevus. A RAF1 FISH break-apart test found a balanced rearrangement pattern in the nevus and an unbalanced pattern in the malignant areas. Wild-type status of NRAS and BRAF was confirmed by NGS techniques. The array-CGH profile displayed copy number alterations commonly found in rhabdomyosarcomas. Despite intensive treatment, widespread metastatic evolution of the melanomatous component was observed.


Subject(s)
Cell Differentiation , Gene Fusion , Melanoma/pathology , Nevus, Pigmented/pathology , Proto-Oncogene Proteins c-raf/genetics , Rhabdomyosarcoma/pathology , Skin Neoplasms/pathology , Child, Preschool , Female , Humans , Melanoma/complications , Melanoma/genetics , Nevus, Pigmented/complications , Nevus, Pigmented/genetics , Rhabdomyosarcoma/complications , Rhabdomyosarcoma/genetics , Skin Neoplasms/complications , Skin Neoplasms/genetics
9.
Virchows Arch ; 474(5): 539-550, 2019 May.
Article in English | MEDLINE | ID: mdl-30756182

ABSTRACT

Recent advances in genomics have improved the molecular classification of cutaneous melanocytic tumors. Among them, deep penetrating nevi (DPN) and plexiform nevi have been linked to joint activation of the MAP kinase and dysregulation of the ß-catenin pathways. Immunohistochemical studies have confirmed cytoplasmic and nuclear expression of ß-catenin and its downstream effector cyclin D1 in these tumors. We assessed nuclear ß-catenin immunohistochemical expression in a large group of DPN as well as in the four most frequent differential diagnoses of DPN: "blue" melanocytic tumors, Spitz tumors, nevoid and SSM melanomas, and pigmented epithelioid melanocytomas (PEM). Nuclear ß-catenin expression was positive in 98/100 DPN and 2/16 of melanomas (one SSM and one nevoid melanoma with a plexiform clone) and was negative in all 30 Spitz, 26 blue, and 6 PEM lesions. In 41% DPN, ß-catenin expression was positive in more than 30% nuclei. No differences were observed in cytoplasmic and nuclear cyclin D1 expression between these tumor groups, suggesting alternate, ß-catenin-independent, activation pathways. We have subsequently studied nuclear ß-catenin expression in a set of 13 tumors with an ambiguous diagnosis, for which DPN was part of the differential diagnosis. The three out of four patients showing canonical DPN mutation profiles were the only ß-catenin-positive cases. We conclude that nuclear ß-catenin expression, independently from CCND1 expression, in a dermal melanocytic tumor is an argument for its classification as DPN. In ambiguous cases and in early combined DPN lesions, this antibody can be helpful as a screening tool. ß-Catenin is also potentially expressed in a subset of malignant melanomas with CTNNB1 mutations.


Subject(s)
Melanoma/pathology , Nevus, Epithelioid and Spindle Cell/metabolism , Skin Neoplasms/pathology , beta Catenin/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Cell Nucleus/pathology , Child , Child, Preschool , Diagnosis, Differential , Female , Humans , Immunohistochemistry/methods , Male , Melanoma/diagnosis , Middle Aged , Mutation/genetics , Nevus, Pigmented/diagnosis , Nevus, Pigmented/pathology , Skin Neoplasms/diagnosis , Young Adult , beta Catenin/genetics , Melanoma, Cutaneous Malignant
10.
J Med Case Rep ; 12(1): 351, 2018 Nov 25.
Article in English | MEDLINE | ID: mdl-30474572

ABSTRACT

BACKGROUND: Renal cell carcinoma represents 3-5% of adult malignant tumors. Metastases are found in 30-40% of patients and brain metastases occurred in more than 10% of them. Despite significant progress in medical treatment, patients with brain metastases still have a limited survival. Cabozantinib, a tyrosine kinase inhibitor directed against vascular endothelial growth factor receptors, was recently registered for the treatment of metastatic renal cell carcinoma. Almost no data are, however, available on patients with brain metastases. CASE PRESENTATION: Case 1 is a 51-year-old man of North African origin; Case 2 is a 55-year-old European man. Case 1 and Case 2 had metastases of renal carcinoma at initial diagnosis and were treated with vascular endothelial growth factor receptors tyrosine kinase inhibitors. Case 1 had clear cell renal carcinoma and underwent nephrectomy; he then received several lines of tyrosine kinase inhibitor directed against vascular endothelial growth factor receptors and the mTor complex. During the second treatment a brain metastasis was diagnosed and treated with radiosurgery with rapid efficacy. Two years later he received nivolumab, an antibody directed against the programmed death-1 and programmed death-ligand 1 complex, but disease progression was observed with the reappearance of the brain metastasis together with neurologic symptoms. Cabozantinib was administered and induced a rapid clinical improvement as well as tumor regression in all sites including his brain. Sequencing of his tumor evidenced a mutation of the MET gene. Case 2 had a papillary renal carcinoma with brain metastases at time of diagnosis. After radiation of the brain tumors, a vascular endothelial growth factor receptor tyrosine kinase inhibitor was administered for 3 years. The disease was under control in all sites except in his brain; several new brain metastases requiring new radiation treatments developed. The disease finally progressed at all metastatic sites including his brain and he had several neurological symptoms. Cabozantinib was administered and rapidly induced a clinical improvement; a further computed tomography scan and brain magnetic resonance imaging showed significant tumor regressions. No MET gene mutation or amplification was observed in the tumor analysis. CONCLUSIONS: These case reports indicate that cabozantinib was able, first, to reach brain tumors and second, to induce significant regressions in renal carcinoma brain metastases that were resistant to radiation as well as to previous systemic vascular endothelial growth factor receptor tyrosine kinase inhibitors.


Subject(s)
Anilides/therapeutic use , Brain Neoplasms/secondary , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Pyridines/therapeutic use , Receptors, Vascular Endothelial Growth Factor/drug effects , Anilides/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Brain Neoplasms/drug therapy , Brain Neoplasms/physiopathology , Carcinoma, Renal Cell/drug therapy , Disease Progression , Humans , Kidney Neoplasms/drug therapy , Male , Middle Aged , Pyridines/pharmacology , Treatment Outcome
11.
Am J Surg Pathol ; 42(3): 382-391, 2018 03.
Article in English | MEDLINE | ID: mdl-29240581

ABSTRACT

We report 5 cases of primary intradermal nodular unpigmented tumors with a melanocytic immunophenotype associated with a novel CRTC1-TRIM11 fusion. Clinically, the cutaneous nodules were slowly growing in 3 women and 2 men (25 to 82 y old, median, 28 y) with no specific topography. Lesion size ranged from 4 to 12 mm (median, 5 mm). The tumors were strictly located in the dermis with a nodular pattern. The cells were arranged in confluent nests and fascicules. Central fibronecrotic areas were present in 2 cases. Cells were medium to large, sometimes multinucleated, and presented a spindled and epithelioid cytology with prominent nucleoli. Cytonuclear atypia was constant, and mitotic activity in hotspot areas ranged from 1 to 5/mm². Immunohistochemistry found a constant positivity with S100, MiTF, and Sox10, and a heterogenous staining by MelanA or HMB45. NTRK1 was strongly positive in 3 cases. In all cases, RNA sequencing found an invariable CRTC1(e1)-TRIM11(e2) fusion, confirmed by fluorescent in situ hybridization techniques with a TRIM11 break-apart probe. In 4/4 cases, nuclear TRIM11 expression was positive by immunohistochemistry. Fluorescent in situ hybridization techniques showed no rearrangement of NTRK1 or EWSR1, and array-comparative genomic hybridization displayed no alteration (1 case) or only a whole chromosome 7 gain (2 cases) when performed. No relapse or metastatic event was observed during follow-up [3 to 72 months (median, 14 mo)]. Cutaneous clear cell sarcoma was the main differential diagnosis. Overlapping morphologic features previously described in primary dermal melanomas and paraganglioma-like melanocytic tumors were present. The CRTC1-TRIM11 fusion appears to be specific of an unpigmented nodular tumor combining a melanocytic phenotype and low-grade tumor behavior.


Subject(s)
Biomarkers, Tumor/genetics , Gene Fusion , Melanocytes , Sarcoma, Clear Cell/genetics , Skin Neoplasms/genetics , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Adult , Aged, 80 and over , Biomarkers, Tumor/analysis , Diagnosis, Differential , Female , Genetic Predisposition to Disease , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Melanocytes/chemistry , Melanocytes/pathology , Middle Aged , Neoplasm Grading , Phenotype , Predictive Value of Tests , Sarcoma, Clear Cell/chemistry , Sarcoma, Clear Cell/pathology , Skin Neoplasms/chemistry , Skin Neoplasms/pathology , Skin Neoplasms/surgery
12.
Nat Commun ; 8(1): 644, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28935960

ABSTRACT

Deep penetrating nevus (DPN) is characterized by enlarged, pigmented melanocytes that extend through the dermis. DPN can be difficult to distinguish from melanoma but rarely displays aggressive biological behavior. Here, we identify a combination of mutations of the ß-catenin and mitogen-activated protein kinase pathways as characteristic of DPN. Mutations of the ß-catenin pathway change the phenotype of a common nevus with BRAF mutation into that of DPN, with increased pigmentation, cell volume and nuclear cyclin D1 levels. Our results suggest that constitutive ß-catenin pathway activation promotes tumorigenesis by overriding dependencies on the microenvironment that constrain proliferation of common nevi. In melanoma that arose from DPN we find additional oncogenic alterations. We identify DPN as an intermediate stage in the step-wise progression from nevus to melanoma. In summary, we delineate specific genetic alterations and their sequential order, information that can assist in the diagnostic classification and grading of these distinctive neoplasms.Deep penetrating nevi (DPN) are unusual melanocytic neoplasms with unknown genetic drivers. Here the authors show that majority of DPN harbor activating mutations in the ß-catenin and the MAP-kinase pathways; this characteristic can help in the classification and grading of these distinctive neoplasms.


Subject(s)
Gene Expression Regulation/physiology , MAP Kinase Signaling System/physiology , Nevus, Intradermal/metabolism , beta Catenin/metabolism , DNA/genetics , DNA/metabolism , Humans , Melanoma/genetics , Melanoma/metabolism , Mutation , Nevus, Intradermal/genetics , beta Catenin/genetics
17.
J Mol Diagn ; 18(2): 205-14, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26752307

ABSTRACT

Personalized medicine has gained increasing importance in clinical oncology, and several clinically important biomarkers are implemented in routine practice. In an effort to guarantee high quality of molecular testing in France, three subsequent external quality assessment rounds were organized at the initiative of the National Cancer Institute between 2012 and 2014. The schemes included clinically relevant biomarkers for metastatic colorectal (KRAS, NRAS, BRAF, PIK3CA, microsatellite instability) and non-small cell lung cancer (EGFR, KRAS, BRAF, PIK3CA, ERBB2), and they represent the first multigene/multicancer studies throughout Europe. In total, 56 laboratories coordinated by 28 regional molecular centers participated in the schemes. Laboratories received formalin-fixed, paraffin-embedded samples and were asked to use routine methods for molecular testing to predict patient response to targeted therapies. They were encouraged to return results within 14 calendar days after sample receipt. Both genotyping and reporting were evaluated separately. During the three external quality assessment rounds, mean genotype scores were all above the preset standard of 90% for all biomarkers. Participants were mainly challenged in case of rare insertions or deletions. Assessment of the written reports showed substantial progress between the external quality assessment schemes on multiple criteria. Several essential elements such as the clinical interpretation of test results and the reason for testing still require improvement by continued external quality assessment education.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Colorectal Neoplasms/genetics , Laboratory Proficiency Testing/standards , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Colorectal Neoplasms/pathology , France , Genetic Testing/standards , Genotyping Techniques/standards , Humans , Lung Neoplasms/pathology , Microsatellite Instability , Time Factors
18.
Am J Surg Pathol ; 40(3): 368-77, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26645730

ABSTRACT

Melanomas associated with blue nevi (MABN) or mimicking cellular blue nevi (MMCBN) represent exceptional variants of malignant cutaneous melanocytic tumors. Uveal and leptomeningeal melanomas frequently have somatic mutations of GNAQ or GNA11, which are believed to be early driver mutations. In uveal melanomas, monosomy 3, linked to the BAP1 gene, is an adverse prognostic factor. We have studied the clinical, histologic, BAP1 expression profile, and molecular data of 11 cases of MABN/MMCBN and 24 cellular blue nevi. Most of the cases of MABN/MMCBN occurred on the scalps of adult patients and presented as rapidly growing nodules, typically >1 cm, often arising at the site of a preexisting melanocytic lesion. The MABN/MMCBN were composed of dense nests of large dermal atypical melanocytes, in some cases lying adjacent to a blue nevus. Four patients developed metastatic disease, and 2 died from their disease. A GNA11 mutation was found in 8/11 cases and a GNAQ mutation in 1 case. Seven of 11 cases showed loss of nuclear BAP1 immunohistochemical (IHC) expression in the malignant component, sparing the adjacent nevus. Array comparative genomic hybridization revealed recurrent deletions of chromosomes 1p, 3p, 4q, 6q, 8p, 16q, and 17q and recurrent gains of chromosomes 6p, 8q, and 21q. The 24 cases of cellular blue nevi frequently occurred on the sacrum, had GNAQ mutations, and showed normal positive IHC staining for BAP1. These results underscore overlapping features in all blue-like malignant melanocytic tumors. Loss of BAP1 IHC expression was restricted to melanomas, including all metastatic cases.


Subject(s)
Biomarkers, Tumor , GTP-Binding Protein alpha Subunits/genetics , Head and Neck Neoplasms/genetics , Melanoma/genetics , Mutation , Nevus, Blue/genetics , Scalp , Skin Neoplasms/genetics , Tumor Suppressor Proteins/analysis , Ubiquitin Thiolesterase/analysis , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biopsy , Comparative Genomic Hybridization , DNA Mutational Analysis , Down-Regulation , Female , Genetic Predisposition to Disease , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Male , Melanoma/enzymology , Melanoma/mortality , Melanoma/pathology , Middle Aged , Nevus, Blue/enzymology , Nevus, Blue/mortality , Nevus, Blue/pathology , Phenotype , Predictive Value of Tests , Retrospective Studies , Skin Neoplasms/enzymology , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Tumor Burden , Young Adult
19.
Histopathology ; 68(2): 279-85, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26033501

ABSTRACT

AIMS: FOXL2 mutation has been consistently identified in adult granulosa cell tumours (A-GCTs). DICER1 mutations have been described predominantly in Sertoli-Leydig cell tumours (SLCTs). The prognostic implication of these mutations remains uncertain, as moderately sized studies have yielded variable outcomes. Our aim was to determine the implications of DICER1 and FOXL2 mutations in 156 ovarian sex cord-stromal tumours (SCSTs). METHODS AND RESULTS: FOXL2 mutations were found in 94% of pathologically confirmed A-GCTs (95/101), in one of eight juvenile granulosa cell tumours (J-GCTs), and in two of 19 SLCTs. DICER1 mutations in the RNase IIIb domain were found in six of 19 SLCTs, two of eight J-GCTs, and one of 12 undifferentiated SCSTs (Und-SCSTs). Comparison of DICER1-mutated SLCTs with DICER1-non-mutated SLCTs showed that patient age at diagnosis was lower and oestrogen receptor expression was more frequent in DICER1-mutated tumours. With a median follow-up of 22 months, two of five DICER1-mutated SLCTs relapsed, in contrast to none of eight DICER1-non-mutated tumours. CONCLUSIONS: Our results suggest that, in contrast to FOXL2 mutations in A-GCT, DICER1 mutations in SLCT might be more useful for prognosis than for diagnosis. However, study of a larger cohort of patients is necessary to establish this. Identification of genetic alterations in SCST offers promising therapeutic options.


Subject(s)
DEAD-box RNA Helicases/genetics , Forkhead Box Protein L2/genetics , Granulosa Cell Tumor/genetics , Ovarian Neoplasms/genetics , Ribonuclease III/genetics , Sertoli-Leydig Cell Tumor/genetics , Sex Cord-Gonadal Stromal Tumors/genetics , Adolescent , Adult , Aged , Female , Granulosa Cell Tumor/diagnosis , Granulosa Cell Tumor/pathology , Humans , Middle Aged , Mutation , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/pathology , Paraffin Embedding , Prognosis , Sertoli-Leydig Cell Tumor/pathology , Sex Cord-Gonadal Stromal Tumors/diagnosis , Sex Cord-Gonadal Stromal Tumors/pathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...