Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Mod Pathol ; 36(6): 100127, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36965331

RESUMO

Epstein-Barr virus (EBV)-associated smooth muscle tumors (EBV-SMTs) are rare smooth muscle neoplasms exclusively associated with immunosuppression, such as in patients with HIV/AIDS, posttransplant, and congenital immunodeficiency. However, the genomic landscape of EBV-SMTs is poorly understood. Leiomyosarcomas harbor genomic instability and multiple recurrent DNA copy number alterations, whereas leiomyomas lack such changes. Thus, this study aimed to fill this knowledge gap by characterizing copy number alterations in EBV-SMTs and correlating this information with clinicopathologic characteristics. Our study investigated and compared the pathologic characteristics and copy number profiles of 9 EBV-SMTs (from 7 post-transplant and AIDS patients), 6 leiomyomas, and 7 leiomyosarcomas, using chromosomal microarray platforms. Our results showed a lower copy number alteration burden in EBV-SMTs and leiomyoma than in leiomyosarcoma. This contrast in the molecular profile between EBV-SMTs and leiomyosarcoma is concordant with the different clinical behaviors and pathologic characteristics exhibited by these tumors. Despite having an overall copy number alteration profile closer to leiomyoma, recurrent copy number gain of oncogenes, such as RUNX1, CCND2, and ETS2, was found in EBV-SMTs. Epigenetic alterations may play an important role in tumorigenesis as recurrent copy number gains were found in histone deacetylases. A gene enrichment analysis also demonstrated enrichment of genes involved in the host response to viral infection, suggesting that the tumor immune microenvironment may play an important role in EBV-SMT tumorigenesis.


Assuntos
Infecções por Vírus Epstein-Barr , Leiomioma , Leiomiossarcoma , Tumor de Músculo Liso , Humanos , Herpesvirus Humano 4/genética , Leiomiossarcoma/genética , Tumor de Músculo Liso/genética , Tumor de Músculo Liso/patologia , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/patologia , Leiomioma/genética , Carcinogênese , Microambiente Tumoral
2.
Genes Chromosomes Cancer ; 60(3): 138-146, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33230916

RESUMO

Uterine epithelioid and myxoid leiomyosarcomas and inflammatory myofibroblastic tumors are rare mesenchymal neoplasms. Next-generation sequencing recently detected novel PGR fusions in uterine epithelioid leiomyosarcomas that demonstrate characteristic rhabdoid and spindled morphology. PLAG1 gene fusions have also been identified in a subset of myxoid leiomyosarcomas and are associated with PLAG1 overexpression. ALK rearrangements underpin the vast majority of uterine inflammatory myofibroblastic tumors, which demonstrate morphologic, and immunohistochemical features similar to those of inflammatory myofibroblastic tumors elsewhere. This review summarizes the morphologic, immunophenotypic, and molecular genetic features of PGR fusion-positive epithelioid leiomyosarcoma, PLAG1 fusion-positive myxoid leiomyosarcoma, and inflammatory myofibroblastic tumors of the uterus.


Assuntos
Proteínas de Ligação a DNA/genética , Receptores de Progesterona/metabolismo , Neoplasias Uterinas/genética , Adulto , Biomarcadores Tumorais/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Fusão Gênica , Rearranjo Gênico , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Pessoa de Meia-Idade , Neoplasias de Tecido Muscular/genética , Neoplasias de Tecido Muscular/metabolismo , Receptores de Progesterona/genética , Tumor de Músculo Liso/genética , Tumor de Músculo Liso/metabolismo , Fatores de Transcrição/genética , Neoplasias Uterinas/metabolismo
3.
Genes Chromosomes Cancer ; 60(3): 129-137, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33099852

RESUMO

Uterine leiomyoma and leiomyosarcoma are located at the ends of the spectrum of smooth muscle lesions. Leiomyosarcoma belongs to the complex genomic sarcomas characterized by complex karyotypes. In contrast, leiomyoma, has a low level of chromosomal complexity. The analysis of genomic profiles of uterine smooth muscle tumors shows that genomic complexity, which is an expression of chromosomal instability, correlates with the metastatic potential and malignity of tumors: the more genetically complex a smooth muscle tumor is, the more malignant is its progression. In uterine tumors with uncertain malignant potential, the assessment of genomic index by CGH array, that is, counting the genomic complexity of a tumor, allows tumors with a risk of recurrence such as leiomyosarcomas to be distinguished from benign tumors like leiomyomas. The prognosis of leiomyosarcoma is poor and the most powerful prognostic factor so far is stage, as the histologic grade is not informative. In the quest to find efficient molecular prognostic factors, the transcriptomic signature CINSARC Nanocind, a mirror of chromosomic complexity and instability, outperforms stage, in both overall and recurrence-free survival. Genomic index and the CINSARC signature will contribute to improving diagnoses, therapeutic strategies, and randomization in future clinical trials. The biological understanding of the links between the CINSARC signature and metastatic mechanisms may lead to the development of new drugs. Furthermore, ctDNA is a promising new technique to detect residual disease and early recurrence.


Assuntos
Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Biomarcadores Tumorais/genética , Hibridização Genômica Comparativa/métodos , Feminino , Expressão Gênica , Genômica , Humanos , Leiomioma/diagnóstico , Leiomioma/genética , Leiomioma/metabolismo , Leiomiossarcoma/diagnóstico , Leiomiossarcoma/genética , Leiomiossarcoma/metabolismo , Recidiva Local de Neoplasia/genética , Neoplasias de Tecido Conjuntivo e de Tecidos Moles/metabolismo , Prognóstico , Sarcoma/diagnóstico , Sarcoma/genética , Sarcoma/metabolismo , Tumor de Músculo Liso/diagnóstico , Tumor de Músculo Liso/genética , Tumor de Músculo Liso/patologia , Transcriptoma , Neoplasias Uterinas/diagnóstico , Neoplasias Uterinas/patologia
4.
Genes Chromosomes Cancer ; 60(3): 210-216, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33099845

RESUMO

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is an autosomal dominant familial syndrome that results from germline mutation in the fumarate hydratase (FH) gene and is associated with an increased risk for smooth muscle tumors of the uterus and skin and renal cell carcinoma. HLRCC associated RCC develop in up to 25% of patients, often presenting in the fourth decade and are high stage, aggressive tumors with poor clinical outcome. Most women with HLRCC develop large and bulky uterine smooth muscle tumors (USMT) in the second to third decade, thus presenting the ideal opportunity for early detection of HLCC to enable timely implementation of surveillance for their RCC risk. However, the concept of screening women with USMT for HLRCC is challenging given that HLRCC is rare but USMT are common. In addition, FH deficiency in USMT can also result from sporadic FH gene aberrations, unrelated to HLRCC, further complicating any potential screening process. Recent studies show that tumor morphology can be used to identify FH deficiency in USMT and thereby direct patients to formal genetic counseling. The low magnification clues of staghorn shaped blood vessels and alveolar pattern should prompt for high magnification examination for eosinophilic cytoplasmic inclusions and oval nuclei containing prominent eosinophilic macronucleoli surrounded by a halo. Additional clues include Schwannoma-like growth and chain-like distribution of the tumor cells. Although immunostains exist for FH and 2SC, their role is limited in the presence of well-developed FH deficient morphology. The prevalence of germline pathogenic mutation in FH among women with USMT with FH deficient morphology is as high as 50% in some studies, with somatic FH mutation accounting for the remainder. Therefore, morphologic evaluation of USMT for features of FH deficiency can serve as a screening tool for HLRCC syndrome by triaging patients to formal hereditary risk assessment.


Assuntos
Carcinoma de Células Renais/genética , Neoplasias Renais/genética , Leiomiomatose/genética , Síndromes Neoplásicas Hereditárias/genética , Neoplasias Cutâneas/genética , Tumor de Músculo Liso/genética , Neoplasias Uterinas/genética , Carcinoma de Células Renais/diagnóstico , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Feminino , Humanos , Neoplasias Renais/diagnóstico , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Leiomiomatose/metabolismo , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Neoplasias de Tecido Conjuntivo e de Tecidos Moles/metabolismo , Síndromes Neoplásicas Hereditárias/metabolismo , Neoplasias Cutâneas/metabolismo , Tumor de Músculo Liso/diagnóstico , Tumor de Músculo Liso/metabolismo , Tumor de Músculo Liso/patologia , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia
5.
Fetal Pediatr Pathol ; 41(6): 1023-1034, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34738861

RESUMO

Background: Primary immunodeficiency (PID) having defects related to lymphocyte cytotoxic pathway or T-cell dysfunction are well known for developing opportunistic infections and Epstein-Barr virus (EBV)-associated diseases. CARMIL2 deficiency is a recently described combined immunodeficiency (CID) disorder characterized by defective CD28-mediated T cell co-stimulation, altered cytoskeletal dynamics, susceptibility to various infections and Epstein Barr Virus smooth muscle tumor (EBV-SMT). Case report: We report a homozygous CARMIL2 pathogenic variant presenting with recurrent infections and EBV associated smooth muscle tumor (SMT) in a child. Conclusion: The present study reports that EBV SMT may occur in a child with CARMIL2 deficiency.


Assuntos
Infecções por Vírus Epstein-Barr , Tumor de Músculo Liso , Criança , Humanos , Herpesvirus Humano 4/genética , Tumor de Músculo Liso/genética , Tumor de Músculo Liso/complicações , Tumor de Músculo Liso/patologia , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/patologia
6.
Int J Mol Sci ; 22(4)2021 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-33557274

RESUMO

Uterine smooth muscle tumors of uncertain malignant potential (STUMPs) represent a heterogeneous group of tumors that cannot be histologically diagnosed as unequivocally benign or malignant. For this reason, many authors are working to obtain a better definition of diagnostic and prognostic criteria. In this work, we analyzed the genomic and epigenomic profile of uterine smooth muscle tumors (USMTs) in order to find similarities and differences between STUMPs, leiomyosarcomas (LMSs) and leiomyomas (LMs), and possibly identify prognostic factors in this group of tumors. Array-CGH data on 23 USMTs demonstrated the presence of a more similar genomic profile between STUMPs and LMSs. Some genes, such as PRKDC and PUM2, with a potential prognostic value, were never previously associated with STUMP. The methylation data appears to be very promising, especially with regards to the divergent profile found in the sample that relapsed, characterized by an overall CGI hypomethylation. Finally, the Gene Ontology analysis highlighted some cancer genes that could play a pivotal role in the unexpected aggressive behavior that can be found in some of these tumors. These genes could prove to be prognostic markers in the future.


Assuntos
Biomarcadores Tumorais/genética , Epigenômica , Regulação Neoplásica da Expressão Gênica , Leiomioma/patologia , Leiomiossarcoma/patologia , Tumor de Músculo Liso/patologia , Neoplasias Uterinas/patologia , Adulto , Idoso , Estudos de Casos e Controles , Metilação de DNA , Feminino , Seguimentos , Genômica , Humanos , Leiomioma/genética , Leiomiossarcoma/genética , Masculino , Pessoa de Meia-Idade , Prognóstico , Tumor de Músculo Liso/genética , Neoplasias Uterinas/genética
7.
Mod Pathol ; 32(11): 1688-1697, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31189997

RESUMO

Uterine myxoid smooth muscle tumors, including myxoid leiomyosarcoma, are rare and their genomic profile has not been fully characterized. With the discovery of uterine sarcomas with ZC3H7B-BCOR fusion and BCOR internal tandem duplications, the differential diagnosis of myxoid smooth muscle lesions is expanding to include molecularly-defined tumors. Thus, we aimed to explore the genomic landscape of myxoid smooth muscle tumor using comprehensive tools. We performed whole exome next-generation sequencing and a pan-sarcoma RNA fusion assay in tumoral paraffin-embedded tissue from nine well-characterized uterine myxoid smooth muscle tumors (seven myxoid leiomyosarcomas and two myxoid smooth muscle tumors of unknown malignant potential). By immunohistochemistry, all tumors were strongly positive for smooth muscle markers and negative for BCOR staining; 4/6 expressed PLAG1. None of the tumors harbored known fusions including ZC3H7B-BCOR, TRPS1-PLAG1, and RAD51B-PLAG1. None harbored exon 15 BCOR internal tandem duplications; however, four tumors contained BCOR internal tandem duplications of unknown significance (mostly intronic). Mutational burden was low (median 3.8 mutations/megabase). DNA damage repair pathway gene mutations, including TP53 and BRCA2, were found. Copy number variation load, inferred from sequencing data, was variable with genomic indexes ranging from 2.2 to 74.7 (median 25.7), with higher indexes in myxoid leiomyosarcomas than myxoid smooth muscle tumors of unknown malignant potential. The absence of clear driver mutations suggests myxoid smooth muscle tumors to be genetically heterogeneous group of tumours and that other genetic (eg., undiscovered translocation) or epigenetic events drive the pathogenesis of uterine myxoid smooth muscle neoplasia.


Assuntos
Tumor de Músculo Liso/genética , Transcriptoma , Neoplasias Uterinas/genética , Adulto , Idoso , Feminino , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Pessoa de Meia-Idade , Hibridização de Ácido Nucleico/métodos
8.
Toxicol Pathol ; 47(5): 577-584, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31064278

RESUMO

The majority of the tumors in the gastrointestinal (GI) tract of rats and mice, with spindle cell morphology, are diagnosed as smooth muscle tumors (SMTs). Similarly, several decades ago human GI tumors with spindle cell morphology were also diagnosed as SMTs. However, later investigations identified most of these tumors in humans as gastrointestinal stromal tumors (GISTs). The GISTs are considered to arise from the interstitial cells of Cajal located throughout the GI tract. Positive immunohistochemical staining with CKIT antibody is a well-accepted diagnostic marker for GISTs in humans. Since there is a considerable overlap between the histomorphology of SMTs and GISTs, it is not possible to distinguish them on hematoxylin and eosin stained sections. As a result, GISTs are not routinely diagnosed in toxicological studies. The current study was designed to evaluate the tumors diagnosed as leiomyoma or leiomyosarcoma in the National Toxicology Program's 2-year bioassays using CKIT, smooth muscle actin, and desmin immunohistochemistry. The results demonstrate that most of the mouse SMTs diagnosed as leiomyoma or leiomyosarcoma are likely GISTs, whereas in rats the tumors are likely SMTs and not GISTs.


Assuntos
Neoplasias Gastrointestinais/patologia , Tumores do Estroma Gastrointestinal/patologia , Trato Gastrointestinal/patologia , Tumor de Músculo Liso/patologia , Animais , Bases de Dados Factuais , Feminino , Neoplasias Gastrointestinais/genética , Tumores do Estroma Gastrointestinal/genética , Imuno-Histoquímica , Leiomioma/genética , Leiomioma/patologia , Leiomiossarcoma/genética , Leiomiossarcoma/patologia , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-kit/genética , Ratos , Tumor de Músculo Liso/genética , Especificidade da Espécie , Testes de Toxicidade
9.
Mod Pathol ; 31(5): 816-828, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29327710

RESUMO

The diagnosis of a uterine smooth muscle lesion is, in the majority of cases, straightforward. However, in a small number of cases, the morphological criteria used in such lesions cannot differentiate with certainty a benign from a malignant lesion and a diagnosis of smooth muscle tumor with uncertain malignant potential (STUMP) is made. Uterine leiomyosarcomas are often easy to diagnose but it is difficult or even impossible to identify a prognostic factor at the moment of the diagnosis with the exception of the stage. We hypothesize, for uterine smooth muscle lesions, that there is a gradient of genomic complexity that correlates to outcome. We first tested this hypothesis on STUMP lesions in a previous study and demonstrated that this 'gray category' could be split according to genomic index into two groups. A benign group, with a low to moderate alteration rate without recurrence and a malignant group, with a highly rearranged profile akin to uterine leiomyosarcomas. Here, we analyzed a large series of 77 uterine smooth muscle lesions (from 76 patients) morphologically classified as 19 leiomyomas, 14 STUMP and 44 leiomyosarcomas with clinicopathological and genomic correlations. We confirmed that genomic index with a cut-off=10 is a predictor of recurrence (P<0.0001) and with a cut-off=35 is a marker for poor overall survival (P=0.035). For the tumors confined to the uterus, stage as a prognostic factor was not useful in survival prediction. At stage I, among the tumors reclassified as molecular leiomyosarcomas (ie, genomic index ≥10), the poor prognostic markers were: 5p gain (overall survival P=0.0008), genomic index at cut-off=35 (overall survival P=0.0193), 13p loss including RB1 (overall survival P=0.0096) and 17p gain including MYOCD gain (overall survival P=0.0425). Based on these findings (and the feasibility of genomic profiling by array-comparative genomic hybridization), genomic index, 5p and 17p gains prognostic value could be evaluated in future prospective chemotherapy trials.


Assuntos
Perfilação da Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos , Tumor de Músculo Liso/diagnóstico , Tumor de Músculo Liso/genética , Neoplasias Uterinas/diagnóstico , Neoplasias Uterinas/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Cromossomos Humanos Par 13/genética , Cromossomos Humanos Par 17/genética , Cromossomos Humanos Par 5/genética , Feminino , Humanos , Leiomioma/diagnóstico , Leiomioma/genética , Leiomioma/patologia , Leiomiossarcoma/diagnóstico , Leiomiossarcoma/genética , Leiomiossarcoma/patologia , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Prognóstico , Tumor de Músculo Liso/patologia , Análise de Sobrevida , Neoplasias Uterinas/patologia
10.
Adv Anat Pathol ; 25(2): 96-105, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28945610

RESUMO

The topic of hereditary gynecologic malignancies readily evokes associations between Lynch syndrome and endometrial adenocarcinoma, or between BRCA mutations and tubo-ovarian serous carcinoma, but other familial associations are less well-known. Two hereditary syndromes are known to be related to uterine mesenchymal tumors: hereditary leiomyomatosis and renal cell carcinoma syndrome and the tuberous sclerosis complex. In the following review, we describe the current literature on these syndromes, summarizing their clinical, morphologic, immunophenotypic, and genetic data. It is possible that the surgical pathologic diagnosis is the first indication of a familial syndrome, thus emphasizing the importance of a pathologist's familiarity with these potentially suggestive lesions.


Assuntos
Síndromes Neoplásicas Hereditárias , Tumor de Músculo Liso/genética , Tumor de Músculo Liso/patologia , Neoplasias Uterinas/genética , Neoplasias Uterinas/patologia , Feminino , Humanos , Leiomiomatose/genética , Leiomiomatose/patologia , Síndromes Neoplásicas Hereditárias/genética , Síndromes Neoplásicas Hereditárias/patologia , Neoplasias de Células Epitelioides Perivasculares/genética , Neoplasias de Células Epitelioides Perivasculares/patologia , Esclerose Tuberosa/complicações , Esclerose Tuberosa/patologia
11.
Cell Mol Biol (Noisy-le-grand) ; 64(5): 7-12, 2018 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-29729689

RESUMO

Uterine smooth muscle tumors constitute a spectrum of neoplasms. Diagnosis of leiomyomas (LMs) is usually straight forwards; however, atypical leiomyomas (ALMs) and smooth muscle tumors of uncertain malignant potential (STUMPs) have overlapping features and need to be distinguished from leiomyosarcoma. To evaluate progesterone receptor (PR), epithelial growth factor receptor (EGF-R), and galectin-3 expression in LMs, ALMs, STUMPs, and leiomyosarcomas and to assess their possible role in differentiating those tumors. Immunoexpression of EGF-R, PR, and galectin-3 were studied in 44 cases of uterine smooth muscle tumors through retrospective study. Studied cases included 20 LM, 9 ALM, 5 STUMP, and 10 leiomyosarcomas. A semiquantitative score was used to evaluate immunohistochemical staining. EGF-R overexpression was detected in leiomyosarcomas while a lack of or reduced EGF-R expression was observed in the nonsarcomatous group (LMs, ALMs, and STUMPs) with a highly significant difference. On the contrary, there was weak or negative PR staining in leiomyosarcomas compared to intense PR expression in the nonsarcomatous group with a highly significant difference. Meanwhile, galectin-3 was downregulated in leiomyosarcomas compared to the nonsarcomatous group with a significant difference. Correlation analysis revealed negative correlation between EGF-R and PR expression with significant statistical results while correlation of galectin-3 with EGF-R and PR showed insignificant statistical results. Immunoexpression of EGF-R, PR, and galectin-3 could help differentiate challenging cases of uterine smooth muscle tumors. Further studies are recommended to investigate interactions between EGF-R, PR, and galectin-3 and to plan new therapeutic strategies.


Assuntos
Biomarcadores Tumorais/genética , Receptores ErbB/genética , Galectina 3/genética , Leiomioma/diagnóstico , Leiomiossarcoma/diagnóstico , Receptores de Progesterona/genética , Tumor de Músculo Liso/diagnóstico , Biomarcadores Tumorais/metabolismo , Proteínas Sanguíneas , Estudos de Casos e Controles , Diagnóstico Diferencial , Receptores ErbB/metabolismo , Feminino , Galectina 3/metabolismo , Galectinas , Humanos , Imuno-Histoquímica , Leiomioma/genética , Leiomioma/metabolismo , Leiomioma/patologia , Leiomiossarcoma/genética , Leiomiossarcoma/metabolismo , Leiomiossarcoma/patologia , Receptores de Progesterona/metabolismo , Estudos Retrospectivos , Tumor de Músculo Liso/genética , Tumor de Músculo Liso/metabolismo , Tumor de Músculo Liso/patologia , Incerteza , Útero/metabolismo , Útero/patologia
12.
Mol Cancer ; 16(1): 101, 2017 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-28592321

RESUMO

Uterine smooth muscle tumors range from benign leiomyomas to malignant leiomyosarcomas. Based on numerous molecular studies, leiomyomas and leiomyosarcomas mostly lack shared mutations and the majority of tumors are believed to develop through distinct mechanisms. To further characterize the molecular variability among uterine smooth muscle tumors, and simultaneously insinuate their potential malignant progression, we examined the frequency of known genetic leiomyoma driver alterations (MED12 mutations, HMGA2 overexpression, biallelic FH inactivation) in 65 conventional leiomyomas, 94 histopathological leiomyoma variants (18 leiomyomas with bizarre nuclei, 22 cellular, 29 highly cellular, and 25 mitotically active leiomyomas), and 51 leiomyosarcomas. Of the 210 tumors analyzed, 107 had mutations in one of the three driver genes. No tumor had more than one mutation confirming that all alterations are mutually exclusive. MED12 mutations were the most common alterations in conventional and mitotically active leiomyomas and leiomyosarcomas, while leiomyomas with bizarre nuclei were most often FH deficient and cellular tumors showed frequent HMGA2 overexpression. Highly cellular leiomyomas displayed the least amount of alterations leaving the majority of tumors with no known driver aberration. Our results indicate that based on the molecular background, histopathological leiomyoma subtypes do not only differ from conventional leiomyomas, but also from each other. The presence of leiomyoma driver alterations in nearly one third of leiomyosarcomas suggests that some tumors arise through leiomyoma precursor lesion or that these mutations provide growth advantage also to highly aggressive cancers. It is clinically relevant to understand the molecular background of various smooth muscle tumor subtypes, as it may lead to improved diagnosis and personalized treatments in the future.


Assuntos
Biomarcadores Tumorais , Fumarato Hidratase/genética , Proteína HMGA2/genética , Complexo Mediador/genética , Tumor de Músculo Liso/genética , Tumor de Músculo Liso/patologia , Neoplasias Uterinas/genética , Neoplasias Uterinas/patologia , Análise Mutacional de DNA , Feminino , Fumarato Hidratase/metabolismo , Expressão Gênica , Proteína HMGA2/metabolismo , Humanos , Complexo Mediador/metabolismo , Mutação , Gradação de Tumores , Estudos Retrospectivos , Tumor de Músculo Liso/metabolismo , Neoplasias Uterinas/metabolismo
14.
Genet Mol Res ; 16(1)2017 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-28340268

RESUMO

We conducted a meta-analysis to examine p16INK4a expression in uterine smooth muscle tumors (USMTs). Although the prognostic value of tumor suppressor p16INK4a has been elucidated in a variety of cancers and precancerous lesions, its role in USMTs is not well established. We searched PubMed, Web of Science, and Embase for publication son p16INK4a expression in USMTs. Strict inclusion and exclusion criteria were imposed. Risk ratios (RRs) with 95% confidence intervals (95%CIs) were calculated to assess the strength of association. Publication bias was estimated using funnel plots and the Egger's regression test. Twelve eligible studies comprising 661 patients were included. Compared with leiomyoma (LM), the figures for the strength of association were as follows: LM variants (RR = 1.53, 95%CI = 1.03-2.27, P = 0.036, random effect); leiomyosarcoma (LMS) (RR = 3.20, 95%CI = 1.68-6.12, P < 0.001, random effect); and smooth muscle tumors of uncertain malignant potential (STUMP) (RR = 2.90, 95%CI = 1.17-7.21, P = 0.022, random effect). p16INK4a expression was significantly higher in LMS than in LM variants (RR = 3.74, 95%CI = 1.96-7.13, P < 0.001, random effect) or STUMP (RR = 1.67, 95%CI = 1.26-2.23, P < 0.001, fixed effect). There was a significant correlation between overexpressed p16INK4a and recurrence rates of USMTs (RR = 1.85, 95%CI = 1.11-3.10, P = 0.019, fixed effect). p16INK4a over expression is a potential biomarker for diagnosing problematic USMTs and it might indicate a worse prognosis. However, there is currently insufficient evidence to assess the prognostic value of p16INK4a in USMTs.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Leiomioma/metabolismo , Leiomiossarcoma/metabolismo , Tumor de Músculo Liso/metabolismo , Neoplasias Uterinas/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Feminino , Genes p16 , Predisposição Genética para Doença , Humanos , Leiomioma/genética , Leiomiossarcoma/genética , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Prognóstico , Tumor de Músculo Liso/genética , Neoplasias Uterinas/genética
15.
Mod Pathol ; 29(5): 500-10, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26892441

RESUMO

Intravenous leiomyomatosis is an unusual smooth muscle neoplasm with quasi-malignant intravascular growth but a histologically banal appearance. Herein, we report expression and molecular cytogenetic analyses of a series of 12 intravenous leiomyomatosis cases to better understand the pathogenesis of intravenous leiomyomatosis. All cases were analyzed for the expression of HMGA2, MDM2, and CDK4 proteins by immunohistochemistry based on our previous finding of der(14)t(12;14)(q14.3;q24) in intravenous leiomyomatosis. Seven of 12 (58%) intravenous leiomyomatosis cases expressed HMGA2, and none expressed MDM2 or CDK4. Colocalization of hybridization signals for probes from the HMGA2 locus (12q14.3) and from 14q24 by interphase fluorescence in situ hybridization (FISH) was detected in a mean of 89.2% of nuclei in HMGA2-positive cases by immunohistochemistry, but in only 12.4% of nuclei in negative cases, indicating an association of HMGA2 expression and this chromosomal rearrangement (P=8.24 × 10(-10)). Four HMGA2-positive cases had greater than two HMGA2 hybridization signals per cell. No cases showed loss of a hybridization signal by interphase FISH for the frequently deleted region of 7q22 in uterine leiomyomata. One intravenous leiomyomatosis case analyzed by array comparative genomic hybridization revealed complex copy number variations. Finally, expression profiling was performed on three intravenous leiomyomatosis cases. Interestingly, hierarchical cluster analysis of the expression profiles revealed segregation of the intravenous leiomyomatosis cases with leiomyosarcoma rather than with myometrium, uterine leiomyoma of the usual histological type, or plexiform leiomyoma. These findings suggest that intravenous leiomyomatosis cases share some molecular cytogenetic characteristics with uterine leiomyoma, and expression profiles similar to that of leiomyosarcoma cases, further supporting their intermediate, quasi-malignant behavior.


Assuntos
Leiomiomatose/patologia , Tumor de Músculo Liso/patologia , Transcriptoma , Neoplasias Vasculares/patologia , Análise Citogenética , Feminino , Perfilação da Expressão Gênica , Humanos , Leiomioma/genética , Leiomioma/patologia , Leiomiomatose/genética , Tumor de Músculo Liso/genética , Neoplasias Uterinas/genética , Neoplasias Uterinas/patologia , Neoplasias Vasculares/genética
16.
Int J Gynecol Pathol ; 35(6): 531-536, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26919578

RESUMO

Individuals with germ line or somatic mutations of fumarate hydratase are predisposed to the development of leiomyomas of the skin and uterus. In this case report, we will describe the uterine smooth muscle tumors in a young patient who was originally diagnosed with ALM from a myomectomy specimen and experienced recurrence many years later. Tumor cells are diffusely immunoreactive for 2SC, suggestive of FH mutations.


Assuntos
Fumarato Hidratase/genética , Leiomioma/patologia , Mutação , Tumor de Músculo Liso/patologia , Neoplasias Uterinas/patologia , Adulto , Biomarcadores Tumorais/análise , Feminino , Predisposição Genética para Doença/genética , Humanos , Leiomioma/genética , Leiomiomatose/genética , Leiomiomatose/patologia , Recidiva Local de Neoplasia/patologia , Tumor de Músculo Liso/genética , Neoplasias Uterinas/genética
17.
Int J Gynecol Pathol ; 35(4): 321-6, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26598981

RESUMO

Uterine tumors with adipocytic differentiation are very uncommon. Mature adipocytes are sometimes seen as an element of smooth muscle neoplasms, more often as lipoleiomyoma, but also in the rare lipoleiomyosarcoma. Exceptional cases have been reported of various subtypes of liposarcoma associated with uterine smooth muscle tumors with or without adipocytic differentiation. We present a case of pleomorphic liposarcoma arising in a lipoleiomyosarcoma of the uterus. Genomic profiling was performed using a validated next generation sequencing panel covering 410 common cancer genes. Alterations were identified in TP53, PTEN, RB1, FAT1 and TERT. The patient's presentation and clinical course as well as the tumor's morphologic, immunohistochemical and molecular genetic findings are reviewed.


Assuntos
Biomarcadores Tumorais/genética , Leiomiossarcoma/patologia , Lipossarcoma/patologia , Tumor de Músculo Liso/patologia , Neoplasias Uterinas/patologia , Idoso , Diferenciação Celular , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Leiomiossarcoma/diagnóstico , Leiomiossarcoma/genética , Lipossarcoma/diagnóstico , Lipossarcoma/genética , Recidiva Local de Neoplasia , Tumor de Músculo Liso/diagnóstico , Tumor de Músculo Liso/genética , Neoplasias Uterinas/diagnóstico , Neoplasias Uterinas/genética , Útero/patologia
18.
Mod Pathol ; 28(7): 1001-10, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25932961

RESUMO

The diagnosis and management of uterine smooth muscle tumors with uncertain malignant potential (STUMP) is often challenging, and genomic data on these lesions as well as on uterine smooth muscle lesions are limited. We tested the hypothesis that genomic profile determination by array-CGH could split STUMP into a benign group with scarce chromosomal alterations akin to leiomyoma and a malignant group with high chromosomal instability akin to leiomyosarcoma. Array-CGH genomic profile analysis was conducted for a series of 29 cases of uterine STUMP. A group of ten uterine leiomyomas and ten uterine leiomyosarcomas served as controls. The mean age was 50 years (range, 24-85) and the follow-up ranged from 12 to 156 months (average 70 months). Since STUMP is a heterogenous group of tumors with genomic profiles that can harbor few to many chromosomal alterations, we compared genomic indices in leiomyomas and leiomyosarcomas and set a genomic index=10 threshold. Tumors with a genomic index <10 were classified as nonrecurring STUMPs and those with a genomic index >10 represented STUMPs with recurrences and unfavorable outcomes. Hence, the genomic index threshold splits the STUMP category into two groups of tumors with different outcomes: a group comparable to leiomyomas and another similar to leiomyosarcomas, but more indolent. In our STUMP series, genomic analysis by array-CGH is an innovative diagnostic tool for problematic smooth muscle uterine lesions, complementary to the morphological evaluation approach. We provide an improved classification method for distinguishing truly malignant tumors from benign lesions within the category of STUMP, especially those with equivocal morphological features.


Assuntos
Leiomioma/diagnóstico , Tumor de Músculo Liso/diagnóstico , Neoplasias Uterinas/diagnóstico , Adulto , Idoso , Idoso de 80 Anos ou mais , Hibridização Genômica Comparativa , Feminino , Humanos , Leiomioma/genética , Leiomioma/patologia , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Tumor de Músculo Liso/genética , Tumor de Músculo Liso/patologia , Neoplasias Uterinas/genética , Neoplasias Uterinas/patologia , Adulto Jovem
20.
Cancer ; 120(20): 3165-77, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24986214

RESUMO

BACKGROUND: Uterine smooth muscle tumors (USMTs) constitute a group of histologic, genetic, and clinical heterogeneous tumors that include at least 6 major histologically defined tumor types: leiomyoma (ULM), mitotically active leiomyoma (MALM), cellular leiomyoma (CLM), atypical leiomyoma (ALM), uncertain malignant potential (STUMP), and leiomyosarcoma (LMS). Apart from ULM and LMS, the nature of these variants is not well defined. METHODS: A total of 167 cases of different USMT variants were collected, reviewed, and diagnostically confirmed based on the World Health Organization and Stanford schemes. These included 38 cases of LMS, 18 cases of STUMP, 42 cases of ALM, 22 cases of CLM, 7 cases of MALM, and 40 cases of ULM. Molecular analysis included selected microRNAs (miRNAs), oncogenes, and tumor suppressors that are highly relevant to USMT. RESULTS: Overall, 49% (17/35) of LMS cases and 7% (1/14) of STUMP cases died due to their USMT, but no deaths were attributed to ALM. miRNA profiling revealed that ALM and LMS shared similar miRNA signatures. P53 mutations and PTEN deletions were significantly higher in LMS, ALM, and STUMP compared with other USMT variants (P < .01). In contrast, MED12 mutations were extremely common in ULM and MALM (> 74%) but were significantly less common (< 15%) in CLM, ALM, STUMP, and LMS (P < .01). CONCLUSION: Six types of USMT have different gene mutation fingerprints. ALM shares many molecular alterations with LMS. Our findings suggest that ALM may be a precursor lesion of LMS or have similar genetic changes during its early stage.


Assuntos
Biomarcadores Tumorais/genética , Leiomioma/genética , Leiomiossarcoma/genética , Tumor de Músculo Liso/genética , Neoplasias Uterinas/genética , Biomarcadores Tumorais/metabolismo , Feminino , Humanos , Leiomioma/metabolismo , Leiomioma/patologia , Leiomiossarcoma/metabolismo , Leiomiossarcoma/patologia , Pessoa de Meia-Idade , Mutação , Tumor de Músculo Liso/metabolismo , Tumor de Músculo Liso/patologia , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA