Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Gynecol Pathol ; 42(2): 207-211, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-35639348

RESUMEN

Vulvar squamous cell carcinoma of the vulva (VSCC) with sarcomatoid features is a rare variant characterized by spindle-cell morphology and occasional heterologous elements. They are difficult to evaluate due to rarity and lack unified nomenclature and histopathologic criteria. Eight cases of sarcomatoid VSCC were retrieved from archival electronic medical records from 2013 to 2021. Patients often presented at a mean age of 78-yr-old at stage FIGO (2018) III or above. The mean greatest diameter was 4.5 cm and mean depth of invasion was 11.5 mm. Spindle cells exhibited fascicular, nested, and cord-like growth patterns, though a haphazard arrangement or a mix of patterns was frequently observed. The sarcomatoid component frequently arose in the context of prior conventional VSCC treated with radiation therapy (n=6, 75% and chemotherapy (n=5, 63%) with latency periods of 5.2 and 5.4 yr, respectively. Associated lesions included differentiated vulvar intraepithelial neoplasia (n=4, 50%), lichen sclerosus (n=5, 63%), and vulvar acanthosis with altered differentiation (n=1, 13%). Immunohistochemistry showed that VSCC with sarcomatoid features aberrantly expressed p53 (n=4, 60%) through diffuse overexpression or null-type patterns. P16 was invariably negative in all cases. These findings suggest that VSCC with sarcomatoid features does not arise from the HPV-related carcinogenic pathway, and that a subset may also arise from the TP53-independent pathway. Recognizing sarcomatoid morphology in VSCC is important since it may confer an elevated risk of nodal metastasis and poorer survival. Larger studies are required to assess the etiology and prognostic implications of VSCC with sarcomatoid features.


Asunto(s)
Carcinoma in Situ , Carcinoma de Células Escamosas , Infecciones por Papillomavirus , Neoplasias de la Vulva , Femenino , Humanos , Anciano , Infecciones por Papillomavirus/patología , Neoplasias de la Vulva/patología , Carcinoma in Situ/patología , Vulva/patología , Carcinoma de Células Escamosas/patología
2.
Int J Gynecol Pathol ; 42(2): 155-158, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-35348475

RESUMEN

Uterine leiomyoma with massive lymphoid infiltration is characterized by a dense lymphoid infiltrate and germinal centers sparing the adjacent myometrium. Only few reports describe this entity and its etiology is unknown. This rare lesion may also exhibit lymphocytic vasculopathy but this has only been reported in the setting of GnRH agonist exposure. We report 2 cases of uterine leiomyoma with massive lymphoid infiltration in which only 1 patient was exposed to GnRH agonists. In both cases, histopathologic analysis showed thick-walled vessels with swollen endothelial cells showing evidence of intramural lymphocytic infiltration, red blood cell extravasation, and medial edema. This constellation of findings represented frank vascular damage and lymphocytic vasculopathy. Our findings suggest that lymphocytic vasculopathy in these lesions may be secondary to factors other than GnRH agonists. Furthermore, both cases showed an angiocentric disposition of germinal centers that has scarcely been alluded to in prior reports. This finding may provide a clue in accurately recognizing leiomyoma with massive lymphoid infiltration. Recognition of this lesion will allow one to avoid mistaking it for mimickers such as inflammatory myofibroblastic tumor, lymphoid malignancies, or other inflammatory processes.


Asunto(s)
Leiomioma , Neoplasias Uterinas , Femenino , Humanos , Neoplasias Uterinas/patología , Células Endoteliales/patología , Leiomioma/patología , Centro Germinal/patología , Hormona Liberadora de Gonadotropina
3.
Semin Cancer Biol ; 77: 110-126, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-32827632

RESUMEN

BRCA1 and BRCA2 are multi-functional proteins and key factors for maintaining genomic stability through their roles in DNA double strand break repair by homologous recombination, rescuing stalled or damaged DNA replication forks, and regulation of cell cycle DNA damage checkpoints. Impairment of any of these critical roles results in genomic instability, a phenotypic hallmark of many cancers including breast and epithelial ovarian carcinomas (EOC). Damaging, usually loss of function germline and somatic variants in BRCA1 and BRCA2, are important drivers of the development, progression, and management of high-grade serous tubo-ovarian carcinoma (HGSOC). However, mutations in these genes render patients particularly sensitive to platinum-based chemotherapy, and to the more innovative targeted therapies with poly-(ADP-ribose) polymerase inhibitors (PARPis) that are targeted to BRCA1/BRCA2 mutation carriers. Here, we reviewed the literature on the responsiveness of BRCA1/2-associated HGSOC to platinum-based chemotherapy and PARPis, and propose mechanisms underlying the frequent development of resistance to these therapeutic agents.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Carcinoma Epitelial de Ovario/genética , Resistencia a Antineoplásicos/genética , Animales , Antineoplásicos/uso terapéutico , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Femenino , Humanos , Mutación , Compuestos de Platino/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico
4.
Am J Hum Genet ; 103(5): 740-751, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388401

RESUMEN

Androgenetic complete hydatidiform moles are human pregnancies with no embryos and affect 1 in every 1,400 pregnancies. They have mostly androgenetic monospermic genomes with all the chromosomes originating from a haploid sperm and no maternal chromosomes. Androgenetic complete hydatidiform moles were described in 1977, but how they occur has remained an open question. We identified bi-allelic deleterious mutations in MEI1, TOP6BL/C11orf80, and REC114, with roles in meiotic double-strand breaks formation in women with recurrent androgenetic complete hydatidiform moles. We investigated the occurrence of androgenesis in Mei1-deficient female mice and discovered that 8% of their oocytes lose all their chromosomes by extruding them with the spindles into the first polar body. We demonstrate that Mei1-/- oocytes are capable of fertilization and 5% produce androgenetic zygotes. Thus, we uncover a meiotic abnormality in mammals and a mechanism for the genesis of androgenetic zygotes that is the extrusion of all maternal chromosomes and their spindles into the first polar body.


Asunto(s)
Andrógenos/genética , Mola Hidatiforme/genética , Mutación/genética , Alelos , Animales , Cromosomas/genética , Femenino , Humanos , Masculino , Mamíferos/genética , Ratones , Ratones Endogámicos C57BL , Oocitos/patología , Embarazo , Cigoto/patología
5.
Mod Pathol ; 34(1): 194-206, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32724153

RESUMEN

TP53 mutations are implicated in the progression of mucinous borderline tumors (MBOT) to mucinous ovarian carcinomas (MOC). Optimized immunohistochemistry (IHC) for TP53 has been established as a proxy for the TP53 mutation status in other ovarian tumor types. We aimed to confirm the ability of TP53 IHC to predict TP53 mutation status in ovarian mucinous tumors and to evaluate the association of TP53 mutation status with survival among patients with MBOT and MOC. Tumor tissue from an initial cohort of 113 women with MBOT/MOC was stained with optimized IHC for TP53 using tissue microarrays (75.2%) or full sections (24.8%) and interpreted using established criteria as normal or abnormal (overexpression, complete absence, or cytoplasmic). Cases were considered concordant if abnormal IHC staining predicted deleterious TP53 mutations. Discordant tissue microarray cases were re-evaluated on full sections and interpretational criteria were refined. The initial cohort was expanded to a total of 165 MBOT and 424 MOC for the examination of the association of survival with TP53 mutation status, assessed either by TP53 IHC and/or sequencing. Initially, 82/113 (72.6%) cases were concordant using the established criteria. Refined criteria for overexpression to account for intratumoral heterogeneity and terminal differentiation improved concordance to 93.8% (106/113). In the expanded cohort, 19.4% (32/165) of MBOT showed evidence for TP53 mutation and this was associated with a higher risk of recurrence, disease-specific death, and all-cause mortality (overall survival: HR = 4.6, 95% CI 1.5-14.3, p = 0.0087). Within MOC, 61.1% (259/424) harbored a TP53 mutation, but this was not associated with survival (overall survival, p = 0.77). TP53 IHC is an accurate proxy for TP53 mutation status with refined interpretation criteria accounting for intratumoral heterogeneity and terminal differentiation in ovarian mucinous tumors. TP53 mutation status is an important biomarker to identify MBOT with a higher risk of mortality.


Asunto(s)
Biomarcadores de Tumor/genética , Análisis Mutacional de ADN , Inmunohistoquímica , Mutación , Neoplasias Quísticas, Mucinosas y Serosas/genética , Neoplasias Ováricas/genética , Proteína p53 Supresora de Tumor/genética , Adulto , Australia , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Quísticas, Mucinosas y Serosas/mortalidad , Neoplasias Quísticas, Mucinosas y Serosas/patología , Neoplasias Quísticas, Mucinosas y Serosas/terapia , América del Norte , Variaciones Dependientes del Observador , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Valor Predictivo de las Pruebas , Pronóstico , Reproducibilidad de los Resultados , Medición de Riesgo , Factores de Riesgo , Análisis de Matrices Tisulares , Reino Unido
6.
Histopathology ; 79(6): 975-988, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34265101

RESUMEN

AIMS: We aimed to characterise a large cohort of non-invasive, human papillomavirus (HPV) and p53-independent verruciform lesions, such as 'vulvar acanthosis with altered differentiation' (VAAD), 'differentiated exophytic vulvar intra-epithelial lesion' (DEVIL) and 'verruciform lichen simplex chronicus' (vLSC). METHODS AND RESULTS: From January 2008 to December 2020 we retrospectively identified 36 eligible patients with verruciform non-invasive lesions (n = 36) and collected clinical, histological and follow-up parameters. Verruciform non-invasive lesions occurred at a median age of 71 years, with a median follow-up of 33.5 months. Clinically, pruritus was only reported in patients with VAAD (n = 3, 21%). Lesion colour was significantly different across categories (P = 0.028). Apart from the histopathological criteria already known to distinguish these entities (hypogranulosis, epithelial pallor and low-magnification architecture), no other significant criteria were discovered and significant overlap was observed, particularly between VAAD and DEVIL. Patients with vLSC trended towards longer survival without recurrence compared to VAAD and DEVIL (P = 0.082), but showed comparable invasion-free survival interval (P = 0.782). Squamous cell carcinomas (SCC) associated with either VAAD, DEVIL or vLSC displayed similar clinical, histopathological and biological parameters. In non-invasive precursor lesions, stromal oedema was associated with invasion (P = 0.015) and remained so upon Cox regression analysis (P = 0.009). CONCLUSION: Our study of HPV and p53 independent non-invasive verruciform lesions of the vulva highlights significant clinical, histopathological and biological overlap between VAAD, DEVIL and vLSC, suggesting that these pre-invasive lesions should be viewed as a spectrum. We also show that stromal features such as oedema might play an import role in progression to invasion.


Asunto(s)
Carcinoma in Situ/patología , Lesiones Precancerosas/patología , Neoplasias de la Vulva/patología , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Humanos , Persona de Mediana Edad , Estudios Retrospectivos , Proteína p53 Supresora de Tumor
7.
Int J Gynecol Pathol ; 40(5): 487-494, 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-33720083

RESUMEN

Uterine sarcomas represent a clinical challenge because of their difficult diagnosis and the poor prognosis of certain subtypes. The aim of this study was to evaluate the expression of the special AT-rich sequence-binding protein 2 (SATB2) in endometrial stromal sarcoma (ESS) and other types of uterine sarcoma by immunohistochemistry. We studied the expression of SATB2 on 71 full tissue sections of endometrial stromal nodule, low-grade ESS, uterine leiomyomas and leiomyosarcoma, undifferentiated uterine sarcoma, adenosarcoma, and carcinosarcoma samples. Nuclear SATB2 expression was then evaluated in an extended sample set using a tissue microarray, including 78 additional uterine tumor samples. Overall, with a cut-off of ≥10% of tumor cell staining as positive, the nuclear SATB2 score was negative in all endometrial stromal nodule samples (n=10) and positive in 83% of low-grade ESS samples (n=29/35), 40% of undifferentiated uterine sarcoma (n=4/10), 13% of leiomyosarcoma (n=2/16), 14% of adenosarcoma (n=3/22), and 8% carcinosarcoma (n=2/25) samples. Furthermore, in ESS patients, direct comparison of nuclear SATB2 scores with clinicopathologic parameters and other reported biomarkers such as progesterone receptor and estrogen receptor showed that nuclear SATB2 was associated with PR expression and a decreased risk of disease-specific death (odds ratio=0.06, 95% confidence interval=0.04-0.81, P=0.04). Our data suggest that SATB2 could be a marker with relative sensitivity (83%) for distinguishing between endometrial stromal nodule and ESS with potential prognostic value.


Asunto(s)
Adenosarcoma/patología , Carcinosarcoma/patología , Leiomioma/patología , Leiomiosarcoma/patología , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Sarcoma Estromático Endometrial/patología , Factores de Transcripción/metabolismo , Neoplasias Uterinas/patología , Adenosarcoma/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinosarcoma/genética , Estudios de Cohortes , Femenino , Humanos , Leiomioma/genética , Leiomiosarcoma/genética , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Persona de Mediana Edad , Estudios Retrospectivos , Sarcoma Estromático Endometrial/genética , Factores de Transcripción/genética , Neoplasias Uterinas/genética , Adulto Joven
8.
Int J Mol Sci ; 22(10)2021 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-34070214

RESUMEN

During tubo-ovarian high-grade serous carcinoma (HGSC) progression, tumoral cells undergo phenotypic changes in their epithelial marker profiles, which are essential for dissemination processes. Here, we set out to determine whether standard epithelial markers can predict HGSC patient prognosis. Levels of E-CADH, KRT7, KRT18, KRT19 were quantified in 18 HGSC cell lines by Western blot and in a Discovery cohort tissue microarray (TMA) (n = 101 patients) using immunofluorescence. E-CADH and KRT7 levels were subsequently analyzed in the TMA of the Canadian Ovarian Experimental Unified Resource cohort (COEUR, n = 1158 patients) and in public datasets. Epithelial marker expression was highly variable in HGSC cell lines and tissues. In the Discovery cohort, high levels of KRT7 and KRT19 were associated with an unfavorable prognosis, whereas high E-CADH expression indicated a better outcome. Expression of KRT7 and E-CADH gave a robust combination to predict overall survival (OS, p = 0.004) and progression free survival (PFS, p = 5.5 × 10-4) by Kaplan-Meier analysis. In the COEUR cohort, the E-CADH-KRT7 signature was a strong independent prognostic biomarker (OS, HR = 1.6, p = 2.9 × 10-4; PFS, HR = 1.3, p = 0.008) and predicted a poor patient response to chemotherapy (p = 1.3 × 10-4). Our results identify a combination of two epithelial markers as highly significant indicators of HGSC patient prognosis and treatment response.


Asunto(s)
Antígenos CD/metabolismo , Biomarcadores de Tumor/metabolismo , Cadherinas/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Neoplasias de las Trompas Uterinas/metabolismo , Queratina-7/metabolismo , Neoplasias Ováricas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Estudios de Cohortes , Cistadenocarcinoma Seroso/mortalidad , Neoplasias de las Trompas Uterinas/mortalidad , Femenino , Humanos , Estimación de Kaplan-Meier , Queratina-19/metabolismo , Neoplasias Pulmonares/metabolismo , Persona de Mediana Edad , Neoplasias Ováricas/mortalidad , Pronóstico , Supervivencia sin Progresión , Neoplasias Gástricas/metabolismo , Vimentina/metabolismo
9.
Mod Pathol ; 33(11): 2361-2377, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32514162

RESUMEN

High-grade serous carcinoma of uterine adnexa (HGSC) is the most frequent histotype of epithelial ovarian cancer and has a poor 5-year survival rate due to late-stage diagnosis and the poor efficacy of standard treatments. Novel biomarkers of cancer outcome are needed to identify new targetable pathways and improve personalized treatments. Cell-surface screening of 26 HGSC cell lines by high-throughput flow cytometry identified junctional adhesion molecule 1 (JAM-A, also known as F11R) as a potential biomarker. Using a multi-labeled immunofluorescent staining coupled with digital image analysis, protein levels of JAM-A were quantified in tissue microarrays from three HGSC patient cohorts: a discovery cohort (n = 101), the Canadian Ovarian Experimental Unified Resource cohort (COEUR, n = 1158), and the Canadian Cancer Trials Group OV16 cohort (n = 267). Low JAM-A level was associated with poorer outcome in the three cohorts by Kaplan-Meier (p = 0.023, p < 0.001, and p = 0.036, respectively) and was an independent marker of shorter survival in the COEUR cohort (HR = 0.517 (0.381-703), p < 0.001). When analyses were restricted to patients treated by taxane-platinum-based chemotherapy, low JAM-A protein expression was associated with poorer responses in the COEUR (p < 0.001) and OV16 cohorts (p = 0.006) by Kaplan-Meier. Decreased JAM-A gene expression was an indicator of poor outcome in gene expression datasets including The Cancer Genome Atlas (n = 606, p = 0.002) and Kaplan-Meier plotter (n = 1816, p = 0.024). Finally, we observed that tumors with decreased JAM-A expression exhibited an enhanced epithelial to mesenchymal transition (EMT) signature. Our results demonstrate that JAM-A expression is a robust prognostic biomarker of HGSC and may be used to discriminate tumors responsive to therapies targeting EMT.


Asunto(s)
Cistadenocarcinoma Seroso/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Molécula A de Adhesión de Unión/metabolismo , Neoplasias Ováricas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Cistadenocarcinoma Seroso/mortalidad , Cistadenocarcinoma Seroso/patología , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Pronóstico , Tasa de Supervivencia
10.
Mod Pathol ; 33(5): 880-892, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31857680

RESUMEN

Hydatidiform mole (HM) is an aberrant human pregnancy characterized by excessive trophoblastic proliferation and abnormal embryonic development. HM has two morphological types, complete (CHM) and partial (PHM), and non-recurrent ones have three genotypic types, androgenetic monospermic, androgenetic dispermic, and triploid dispermic. Most available studies on risk factors predisposing to different types of HM and their malignant transformation mainly suffer from the lack of comprehensive genotypic analysis of large cohorts of molar tissues combined with accurate postmolar hCG follow-up. Moreover, 10-20% of patients with one HM have at least one non-molar miscarriage, which is higher than the frequency of two pregnancy losses in the general population (2-5%), suggesting a common genetic susceptibility to HM and miscarriages. However, the underlying causes of the miscarriages in these patients are unknown. Here, we comprehensively analyzed 204 HM, mostly from patients referred to the Quebec Registry of Trophoblastic Diseases and for which postmolar hCG monitoring is available, and 30 of their non-molar miscarriages. We revisited the risk of maternal age and neoplastic transformation across the different HM genotypic categories and investigated the presence of chromosomal abnormalities in their non-molar miscarriages. We confirm that androgenetic CHM is more prone to gestational trophoblastic neoplasia (GTN) than triploid dispermic PHM, and androgenetic dispermic CHM is more prone to high-risk GTN and choriocarcinoma (CC) than androgenetic monospermic CHM. We also confirm the association between increased maternal age and androgenetic CHM and their malignancies. Most importantly, we demonstrate for the first time that patients with an HM and miscarriages are at higher risk for aneuploid miscarriages [83.3%, 95% confidence interval (CI): 0.653-0.944] than women with sporadic (51.5%, 95% CI: 50.3-52.7%, p value = 0.0003828) or recurrent miscarriages (43.8%, 95% CI: 40.7-47.0%, p value = 0.00002). Our data suggest common genetic female germline defects predisposing to HM and aneuploid non-molar miscarriages in some patients.


Asunto(s)
Mola Hidatiforme/genética , Neoplasias Uterinas/genética , Aborto Habitual/genética , Adulto , Femenino , Genotipo , Humanos , Edad Materna , Persona de Mediana Edad , Embarazo , Factores de Riesgo
12.
Gynecol Oncol ; 156(2): 377-386, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31753525

RESUMEN

BACKGROUND: Carriers of pathogenic variants in both BRCA1 and BRCA2 genes as a double mutation (BRCA1/2 DM) have been rarely reported in women with epithelial ovarian cancer (EOC). METHODS: We reviewed the English literature and interrogated three repositories reporting EOC patients carrying BRCA1/2 DM. The clinicopathological parameters of 36 EOC patients carrying germline BRCA1/2 DM were compared to high-grade serous EOC women of the COEUR cohort with known germline BRCA1/BRCA2 mutation carrier status (n = 376 non-carriers, n = 65 BRCA1 and n = 38 BRCA2). Clinicopathological parameters evaluated were age at diagnosis, stage of disease, loss of heterozygosity, type of mutation, immunohistochemistry profile, progression occurrence and survival. RESULTS: Median age at diagnosis of BRCA1/2 DM patients was 51.9 years, similar to BRCA1 mutation carriers (49.7 years, p = .58) and younger than BRCA2 mutation carriers (58.1 years, p = .02). Most patients were diagnosed at advanced stage (III-IV; 82%) and were carriers of founder/frequent mutations (69%). Tissue immunostainings revealed no progesterone receptor expression and low intraepithelial inflammation. The 5-year survival rate (60%) was significantly lower than that of BRCA2 mutation carriers (76%, p = .03) but not of BRCA1 mutation carriers (51%, p = .37). CONCLUSIONS: Our data suggests some co-dominant effect of both mutations but the outcome of these patients more closely resembled that of BRCA1 mutation carriers with poor prognosis factors.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Carcinoma Epitelial de Ovario/genética , Neoplasias Ováricas/genética , Adulto , Anciano , Carcinoma Epitelial de Ovario/patología , Femenino , Genes BRCA1 , Genes BRCA2 , Mutación de Línea Germinal , Heterocigoto , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/patología , Receptores de Progesterona/biosíntesis , Receptores de Progesterona/genética
13.
Gynecol Oncol ; 157(1): 36-45, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32001076

RESUMEN

OBJECTIVE: Patients with advanced low-grade serous carcinoma (LGSC) have poor long-term survival rates. As a rare histotype, there are uncertainties regarding the use of current therapies. Thus, we studied practice patterns and treatment outcomes as part of a national initiative to better understand and improve the care of women with advanced LGSC. METHODS: This retrospective cohort study was conducted in 5 Canadian referral institutions from 2000 to 2016. Data collection and pathology reporting were standardized. Outcome measures included overall survival (OS), progression-free survival (PFS), progression-free intervals (PFI), and time to next treatment (TTNT). Cox regression analysis was used to evaluate the effects of clinical and pathologic factors on outcomes and prognosis. RESULTS: There were 134 patients (stage II-IV) with a median follow-up of 32.4 months (range 1.6-228). Four primary treatments were compared across institutions: 1) surgery followed by chemotherapy (56%), 2) neoadjuvant chemotherapy (NACT) followed by surgery (27%), 3) surgery alone (9%), and 4) surgery followed by anti-hormone therapy (4%). Primary platinum/paclitaxel chemotherapy was used in 81%. Patients treated with NACT had worse PFS. Multivariable Cox regression analysis identified lesser residual disease, younger age, and primary peritoneal origin as variables significantly associated with better OS/PFS (p < 0.03). One institution had significantly better PFS than the others (p = 0.025), but this finding could be related to a higher frequency of primary peritoneal LGSC. PFI and TTNT intervals in patients with relapsed disease were not significantly different after the first relapse irrespective of treatment type. CONCLUSIONS: There are notable differences in practice patterns across Canada. This underscores the need for ongoing strategies to measure, evaluate and achieve optimal patient outcomes for women with advanced LGSC.


Asunto(s)
Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/cirugía , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/cirugía , Pautas de la Práctica en Medicina , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carboplatino/administración & dosificación , Cisplatino/administración & dosificación , Estudios de Cohortes , Cistadenocarcinoma Seroso/patología , Femenino , Humanos , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias Ováricas/patología , Paclitaxel/administración & dosificación , Supervivencia sin Progresión , Estudios Retrospectivos , Tasa de Supervivencia , Resultado del Tratamiento
14.
Int J Gynecol Pathol ; 39(4): 362-366, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31033798

RESUMEN

Leiomyosarcoma (LMS) is a malignant mesenchymal neoplasm showing smooth muscle differentiation. Uterine LMS is more frequent that nonuterine LMS, and represents 1% of all malignant neoplasms of the uterus. Pleomorphic undifferentiated uterine sarcoma is a rare entity, and is defined by high-grade sarcoma histology with loss of muscular markers. Several cases of pleomorphic undifferentiated uterine sarcoma have been reported in the literature, with worse clinical outcome when compared with conventional LMS. Here we report the first case of a pleomorphic undifferentiated uterine sarcoma in association with LMS in a 33 yr old woman. The patient presented clinically with recurrent vaginal bleeding and suspicion of a trophoblastic tumor. Ancillary testing revealed moderately elevated beta-hCG (49.7 U/L) and no metastatic disease on imaging. Gross examination of the hysterectomy specimen revealed a large heterogenous necrotic uterine mass infiltrating <50% of the myometrium. Microscopic evaluation showed pleomorphic undifferentiated uterine sarcoma adjacent to a nodule of leiomyoma with bizarre nuclei, with loss of myogenic markers in the high grade component. Other findings included a foci of conventional LMS, and diffuse uterine leiomyomatosis. Although beta-hCG dropped to normal levels during follow-up, the patient developed metastatic lesions to the lung at 6 mo postop. Initial elevation of beta-hCG may have correlated with the aggressive histology of the tumor, as reported by some groups previously. Recognition of pleomorphic undifferentiated uterine sarcoma and its distinction from conventional LMS is essential for patient prognosis and management.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Leiomioma/diagnóstico , Leiomiosarcoma/diagnóstico , Sarcoma/diagnóstico , Neoplasias Uterinas/diagnóstico , Adulto , Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Diagnóstico Diferencial , Femenino , Humanos , Inmunohistoquímica , Leiomioma/patología , Leiomiosarcoma/patología , Miometrio/patología , Pronóstico , Sarcoma/patología , Neoplasias Uterinas/patología , Útero/patología
15.
Int J Gynecol Pathol ; 39(2): 128-135, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30789501

RESUMEN

The origin of serous endometrial intraepithelial carcinoma (SEIC) is debated, due to its premalignant and independently malignant nature. It often arises next to endometrial serous carcinoma (ESC), with a propensity for polypoid growth. We aimed to better characterize this discrepancy by analyzing the clinical, histologic, and immunohistochemical features of polypoid carcinoma associated with SEIC (P-SEIC), and compared them with usual endometrial serous carcinoma without SEIC (UESC). Consecutive patients with P-SEIC were recruited and compared with UESC controls from our institutional research center. Clinical, histologic, and immunohistochemical (IHC, ER, PR, P53, Napsin-A, WT1, P16) were analyzed. BRCA testing results and familial history were also extracted from clinical databases. Welch T test, Pearson χ, and Fisher exact test were performed in SPSS version 23. A total of 37 P-SEIC and 25 UESC were the basis of a case-control study. P-SEIC was associated with more bilateral ovarian involvement (P=0.026), yet showed lower rates of myometrial invasion (P=0.002). P-SEIC showed a statistically different IHC profile: p53+, p16+, ER+, PR+, and WT-1+, and high rates of Napsin-A, while UESC was p53+, p16+, WT-1-, Napsin-A-, with lower rates of ER and PR. We also identified 2 patients who received prophylactic salpingo-oophorectomy for BRCA mutations and who subsequently developed P-SEIC with its unique IHC pattern. Our results suggest different underlying expression profiles and possibly diverging molecular signatures between both P-SEIC and UESC. If confirmed in further molecular studies, it could lead to a distinct molecular subclass.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma in Situ/patología , Cistadenocarcinoma Seroso/patología , Neoplasias Endometriales/patología , Anciano , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Estudios de Casos y Controles , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Pólipos
16.
Mod Pathol ; 31(7): 1116-1130, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29463882

RESUMEN

Hydatidiform mole is an aberrant human pregnancy characterized by early embryonic arrest and excessive trophoblastic proliferation. Recurrent hydatidiform moles are defined by the occurrence of at least two hydatidiform moles in the same patient. Fifty to eighty percent of patients with recurrent hydatidiform moles have biallelic pathogenic variants in NLRP7 or KHDC3L. However, in the remaining patients, the genotypic types of the moles are unknown. We characterized 80 new hydatidiform mole tissues, 57 of which were from patients with no mutations in the known genes, and we reviewed the genotypes of a total of 123 molar tissues. We also reviewed mutation analysis in 113 patients with recurrent hydatidiform moles. While all hydatidiform moles from patients with biallelic NLRP7 or KHDC3L mutations are diploid biparental, we demonstrate that those from patients without mutations are highly heterogeneous and only a small minority of them are diploid biparental (8%). The other mechanisms that were found to recur in patients without mutations are diploid androgenetic monospermic (24%) and triploid dispermic (32%); the remaining hydatidiform moles were misdiagnosed as moles due to errors in the analyses and/or their unusual mechanisms. We compared three parameters of genetic susceptibility in patients with and without mutations and show that patients without mutations are mostly from non-familial cases, have fewer reproductive losses, and more live births. Our data demonstrate that patients with recurrent hydatidiform moles and no mutations in the known genes are, in general, different from those with mutations; they have a milder genetic susceptibility and/or a multifactorial etiology underlying their recurrent hydatidiform moles. Categorizing these patients according to the genotypic types of their recurrent hydatidiform moles may facilitate the identification of novel genes for this entity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Mola Hidatiforme/genética , Neoplasias Primarias Secundarias/genética , Proteínas/genética , Neoplasias Uterinas/genética , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Embarazo
17.
BMC Cancer ; 18(1): 347, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29587661

RESUMEN

BACKGROUND: Ovarian carcinoma is the most lethal gynecological malignancy due to early dissemination and acquired resistance to platinum-based chemotherapy. Reliable markers that are independent and complementary to clinical parameters are needed to improve the management of patients with this disease. The Canadian Ovarian Experimental Unified Resource (COEUR) provides researchers with biological material and associated clinical data to conduct biomarker validation studies. Using standards defined by the Canadian Tissue Repository Network (CTRNet), we have previously demonstrated the quality of the biological material from this resource. Here we describe the clinical characteristics of the COEUR cohort. METHODS: With support from 12 Canadian ovarian cancer biobanks in Canada, we created a central retrospective cohort comprised of more than 2000 patient tissue samples with associated clinical data, including 1246 high-grade serous, 102 low-grade serous, 295 endometrioid, 259 clear cell and 89 mucinous carcinoma histotypes. A two-step reclassification process was applied to assure contemporary histological classification (histotyping). For each histotypes individually, we evaluated the association between the known clinico-pathological parameters (stage, cytoreduction, chemotherapy treatment, BRCA1 and BRCA2 mutation) and patient outcome by using Kaplan-Meier and Cox proportional hazard regression analyses. RESULTS: The median follow-up time of the cohort was 45 months and the 5-year survival rate for patients with high-grade serous carcinomas was 34%, in contrast to endometrioid carcinomas with 80% at 5 years. Survival profiles differed by histotype when stratified by stage or cytoreduction. Women with mucinous or clear cell carcinomas at advanced stage or with non-optimally debulked disease had the worst outcomes. In high-grade serous carcinoma, we observed significant association with longer survival in women harboring BRCA1 or BRCA2 mutation as compared to patients without detectable mutation. CONCLUSIONS: Our results show the expected survival rates, as compared with current literature, in each histotype suggesting that the cohort is an unbiased representation of the five major histotypes. COEUR, a one stop comprehensive biorepository, has collected mature outcome data and relevant clinical data in a comprehensive manner allowing stratified analysis.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Ováricas/diagnóstico , Anciano , Bancos de Muestras Biológicas , Canadá , Estudios de Cohortes , Femenino , Genes BRCA1 , Genes BRCA2 , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/mortalidad , Pronóstico , Modelos de Riesgos Proporcionales
20.
Int J Gynecol Pathol ; 35(1): 30-2, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26166717

RESUMEN

Numerous histologic variants of uterine leiomyomas have been described. The main interest in recognizing these variants is differentiating them from leiomyosarcoma. Osteoclast-like giant cells (OLGC) have been described in association with leiomyosarcoma but to our knowledge, never with leiomyoma. We here report the case of a 58-year-old woman who underwent an elective total hysterectomy with bilateral salpingo-oophorectomy and bilateral pelvic lymphadenectomy for endometrial atypical complex hyperplasia. Multiple typical uterine leiomyoma were identified. One of them showed numerous OLGC admixed with fascicules of bland smooth muscle cells. No atypical features were identified in multiple sections of this otherwise classic uterine leiomyoma. The OLGC showed strong positivity for CD68. The patient, on follow-up, did not show any evidence of recurrent or metastatic disease. This unusual finding expands the morphologic spectrum of uterine leiomyomas. When confronted with a uterine smooth muscle cell tumor with an OLGC component, it is important to search for atypical features diagnostic of leiomyosarcoma.


Asunto(s)
Leiomioma/patología , Leiomiosarcoma/patología , Neoplasias Uterinas/patología , Diagnóstico Diferencial , Femenino , Células Gigantes/patología , Humanos , Histerectomía , Leiomioma/cirugía , Persona de Mediana Edad , Osteoclastos/patología , Neoplasias Uterinas/cirugía , Útero/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA