Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 437
Filtrar
1.
Exp Dermatol ; 32(10): 1734-1743, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37486173

RESUMEN

Incorrect and delayed diagnosis of vulvar high-grade squamous intraepithelial neoplasia (vHSIL) and lichen sclerosus (LS) increases malignant progression risks and negatively impacts prognosis and quality of life. There is a need to improve diagnosis and monitoring. Reflectance confocal microscopy is a non-invasive imaging tool that visualizes skin structures at cellular resolution. The objectives were to explore feasibility and patient acceptability of vulvar RCM imaging and to identify RCM characteristics that are discriminative for vulvar HSIL and LS. This was a prospective, cross-sectional, observational clinical trial in patients with vHSIL and LS compared to healthy volunteers. RCM images and vulvar tissue samples were obtained. Five (5) patients with vHSIL, 10 patients with LS and 10 healthy volunteers were enrolled. In total, 100 image series of vulvar skin were obtained, including lesional and nonlesional sites. The RCM technique was considered acceptable for application by patients and healthy controls. Healthy vulvar skin was characterized by a homogenous, normal honeycomb patterned epidermis and a clear epidermal-dermal junctions. Vulvar HSIL and LS displayed an atypical honeycomb pattern of the epidermis and lymphocytic influx with presence of melanophages. Distinct features specifically observed in LS included the presence of hyalinised vessels and sclerotic areas in the dermis. RCM is a non-invasive imaging technique that is feasible and clinically acceptable to apply on vulvar skin, both in patients with premalignant lesions and healthy controls. Recognition and validation of disease-specific characteristics could make reflectance confocal microscopy a clinical tool to non-invasively aid identification of vulvar premalignancies.


Asunto(s)
Carcinoma in Situ , Liquen Escleroso y Atrófico , Neoplasias Cutáneas , Neoplasias de la Vulva , Femenino , Humanos , Liquen Escleroso y Atrófico/diagnóstico por imagen , Liquen Escleroso y Atrófico/patología , Estudios Transversales , Voluntarios Sanos , Estudios Prospectivos , Calidad de Vida , Neoplasias de la Vulva/diagnóstico por imagen , Neoplasias de la Vulva/patología , Neoplasias Cutáneas/patología , Carcinoma in Situ/química , Carcinoma in Situ/patología , Microscopía Confocal
2.
Hum Pathol ; 136: 56-62, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36997033

RESUMEN

Urothelial carcinoma in situ (uCIS) is typically recognized by overtly malignant cells with characteristic nuclear features; multiple histologic patterns have been described. A rare "overriding" pattern, in which uCIS tumor cells extend on top of normal urothelium, has previously been mentioned in the literature, but not well described. Herein, we report 3 cases of uCIS with "overriding" features. Detailed morphologic evaluation revealed somewhat subtle cytologic atypia: variably enlarged hyperchromatic nuclei and scattered mitotic figures but with abundant cytoplasm and limited to superficial urothelium. Immunohistochemical (IHC) analysis showed a distinctive diffuse positive aberrant p53 pattern, limited to the atypical surface urothelial cells; these cells also showed CK20+, CD44-, and increased Ki-67. In 2 cases, there was a history of urothelial carcinoma and adjacent conventional uCIS. In the third case, the "overriding" pattern was the first presentation of urothelial carcinoma; therefore, next-generation sequencing molecular testing was also performed, revealing pathogenic mutations in TERTp, TP53, and CDKN1a to further support neoplasia. Notably, the "overriding" pattern mimicked umbrella cells, which normally line surface urothelium, can have abundant cytoplasm and more variation in nuclear and cell size and shape, and show CK20+ IHC. We therefore also evaluated umbrella cell IHC patterns in adjacent benign/reactive urothelium, which showed CK20+, CD44-, p53 wild-type, and very low Ki-67 (3/3). We also reviewed 32 cases of normal/reactive urothelium: all showed p53 wild-type IHC in the umbrella cell layer (32/32). In conclusion, caution is warranted to avoid overdiagnosis of usual umbrella cells as CIS; however, "overriding" uCIS should be recognized, may have morphologic features that fall short of the diagnostic threshold of conventional CIS, and requires further study.


Asunto(s)
Carcinoma in Situ , Carcinoma de Células Transicionales , Neoplasias de la Vejiga Urinaria , Humanos , Neoplasias de la Vejiga Urinaria/química , Carcinoma de Células Transicionales/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/análisis , Antígeno Ki-67/análisis , Carcinoma in Situ/diagnóstico , Carcinoma in Situ/genética , Carcinoma in Situ/química , Urotelio/patología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/análisis
3.
Diagn Pathol ; 16(1): 95, 2021 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-34689808

RESUMEN

BACKGROUND: The precursors of clear cell endometrial carcinoma (CC-EC) are still undefined. Here, we deal with the diagnostic issues related to CC-EC precursors by presenting a morphological, immunophenotypical and molecular study of two representative cases and discussing the relevant literature. CASE PRESENTATION: Our and previous cases suggest that clear cell endometrial intraepithelial carcinoma (CC-EIC) is a real entity, which may be distinguished from metaplastic/reactive changes and from its serous counterpart. CC-EIC appears associated with atrophic polyps and may be diagnosed based on morphological and immunophenotypical features of CC-EC in the absence of invasive disease. We described a p53-mutant putative precursor characterized by high-grade nuclei in the absence of other distinctive features. Two putative low-grade precursors resembled atypical tubal metaplasia and endometrial intraepithelial neoplasia, although immunohistochemistry could not support their relationship with CC-EC. CONCLUSIONS: In conclusion, pathologists should be aware of the existence of CC-EIC, since its correct diagnosis may be crucial for a correct patient management. Although several putative earlier precursors have been described, they does not show univocal features that allow their recognition in the common practice. Further studies are necessary in this field.


Asunto(s)
Carcinoma in Situ/patología , Neoplasias Endometriales/patología , Neoplasias Quísticas, Mucinosas y Serosas/patología , Anciano , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Biopsia , Carcinoma in Situ/química , Carcinoma in Situ/genética , Análisis Mutacional de ADN , Neoplasias Endometriales/química , Neoplasias Endometriales/genética , Femenino , Humanos , Inmunohistoquímica , Metaplasia , Mutación , Clasificación del Tumor , Neoplasias Quísticas, Mucinosas y Serosas/química , Neoplasias Quísticas, Mucinosas y Serosas/genética , Valor Predictivo de las Pruebas , Proteína p53 Supresora de Tumor/genética
4.
Am J Surg Pathol ; 45(11): 1573-1578, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34231547

RESUMEN

p16 is the most useful diagnostic marker for human papillomavirus (HPV)-associated anogenital lesions. In the cervix, the pattern of p16 immunoreactivity generally correlates with lesion severity. p16 expression in anal intraepithelial neoplasia (AIN) is far less studied. Whether such correlation holds true has to be determined. We correlated the degree and pattern of p16 immunohistochemistry (IHC) results with morphologic diagnoses of 1000 anal squamous and transitional zone biopsy specimens. Using the Lower Anogenital Squamous Terminology criteria, p16 IHC results were classified as block staining, partial staining, or negative. Among 150 samples without morphologic evidence of AIN, p16 was negative in 85% and partial staining in 15%. AIN 1 (n=400) revealed diverse results: 28% negative, 35% partial, and 37% block staining. Among AIN 2 (n=298), 89% were block, 9% partial staining, and 2% negative. AIN 3 (n=152) revealed block (95%) or partial staining (5%). For the detection of AIN 2/3, p16 block staining yielded 91% sensitivity, 73% specificity, 80% positive predictive value, 91% negative predictive value, and a Youden Index of 0.64. Combining block staining and partial staining slightly increased sensitivity (99%) and negative predictive value (98%), but significantly decreased specificity (43%), positive predictive value (59%) and Youden Index (0.42, P<0.001). As with the cervix, p16 immunoreactivity correlates with morphologic diagnoses of AIN. Block staining offers the optimal diagnostic value for AIN 2/3. Caution is required since AIN 1 frequently exhibits block staining; the prognostic value of p16 warrants further investigation.


Asunto(s)
Neoplasias del Ano/química , Biomarcadores de Tumor/análisis , Carcinoma in Situ/química , Inhibidor p16 de la Quinasa Dependiente de Ciclina/análisis , Inmunohistoquímica , Papillomaviridae/aislamiento & purificación , Infecciones por Papillomavirus/virología , Lesiones Precancerosas/metabolismo , Lesiones Intraepiteliales Escamosas/metabolismo , Neoplasias del Ano/patología , Neoplasias del Ano/virología , Biopsia , Carcinoma in Situ/patología , Carcinoma in Situ/virología , Bases de Datos Factuales , Humanos , Hibridación in Situ , Masculino , Clasificación del Tumor , Papillomaviridae/genética , Lesiones Precancerosas/patología , Lesiones Precancerosas/virología , Valor Predictivo de las Pruebas , ARN Viral/genética , Lesiones Intraepiteliales Escamosas/patología , Lesiones Intraepiteliales Escamosas/virología
5.
Virchows Arch ; 479(2): 325-335, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33650041

RESUMEN

Reliable factors predicting the disease course of non-muscle-invasive bladder cancer (NMIBC) with carcinoma in situ (CIS) are unavailable. Molecular subtypes have potential for prognostic stratification of muscle-invasive bladder cancer, while their value for CIS patients is unknown. Here, the prognostic impact of both clinico-pathological parameters, including CIS focality, and immunohistochemistry-based surrogate subtypes was analyzed in a cohort of high-risk NMIBC patients with CIS. In 128 high-risk NMIBC patients with CIS, luminal (KRT20, GATA3, ERBB2) and basal (KRT5/6, KRT14) surrogate markers as well as p53 were analyzed in 213-231 biopsies. To study inter-lesional heterogeneity of CIS, marker expression in independent CIS biopsies from different bladder localizations was analyzed. Clinico-pathological parameters and surrogate subtypes were correlated with recurrence-free (RFS), progression-free (PFS), cancer-specific (CSS), and overall survival (OS). Forty-six and 30% of CIS patients exhibited a luminal-like (KRT20-positive, KRT5/6-negative) and a null phenotype (KRT20-negative, KRT5/6-negative), respectively. A basal-like subtype (KRT20-negative, KRT5/6-positive) was not observed. A significant degree of inter-lesional CIS heterogeneity was noted, reflected by 23% of patients showing a mixed subtype. Neither CIS surrogate subtype nor CIS focality was associated with patient outcome. Patient age and smoking status were the only potentially independent prognostic factors predicting RFS, PFS, OS, and PFS, respectively. In conclusion, further clarification of heterogeneity of surrogate subtypes in HR NMIBC and their prognostic value is of importance with regard to potential implementation of molecular subtyping into clinical routine. The potential prognostic usefulness of patient age and smoking status for high-risk NMIBC patients with CIS needs further validation.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma in Situ/química , Inmunohistoquímica , Neoplasias de la Vejiga Urinaria/química , Urotelio/química , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Carcinoma in Situ/mortalidad , Carcinoma in Situ/patología , Carcinoma in Situ/terapia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Recurrencia Local de Neoplasia , Valor Predictivo de las Pruebas , Supervivencia sin Progresión , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Fumar/efectos adversos , Análisis de Matrices Tisulares , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/terapia , Urotelio/patología
6.
Virchows Arch ; 479(2): 305-315, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33682013

RESUMEN

Differentiated vulvar intraepithelial neoplasia (dVIN) is a premalignant lesion that is known to progress rapidly to invasive carcinoma. Accurate histological diagnosis is therefore crucial to allow appropriate treatment. To identify reliable diagnostic features, we evaluated the inter-observer agreement in the histological assessment of dVIN, among a bi-national, multi-institutional group of pathologists. Two investigators from Erasmus MC selected 36 hematoxylin-eosin-stained glass slides of dVIN and no-dysplasia, and prepared a list of 15 histological features of dVIN. Nine participating pathologists (i) diagnosed each slide as dVIN or no-dysplasia, (ii) indicated which features they used for the diagnosis, and (iii) rated these features in terms of their diagnostic usefulness. Diagnoses rendered by > 50% participants were taken as the consensus (gold standard). p53-immunohistochemistry (IHC) was performed for all cases, and the expression patterns were correlated with the consensus diagnoses. Kappa (ĸ)-statistics were computed to measure inter-observer agreements, and concordance of the p53-IHC patterns with the consensus diagnoses. For the diagnosis of dVIN, overall agreement was moderate (ĸ = 0.42), and pair-wise agreements ranged from slight (ĸ = 0.10) to substantial (ĸ = 0.73). Based on the levels of agreement and ratings of usefulness, the most helpful diagnostic features were parakeratosis, cobblestone appearance, chromatin abnormality, angulated nuclei, atypia discernable under × 100, and altered cellular alignment. p53-IHC patterns showed substantial concordance (ĸ = 0.67) with the consensus diagnoses. Histological interpretation of dVIN remains challenging with suboptimal inter-observer agreement. We identified the histological features that may facilitate the diagnosis of dVIN. For cases with a histological suspicion of dVIN, consensus-based pathological evaluation may improve the reliability of the diagnosis.


Asunto(s)
Carcinoma in Situ/patología , Diferenciación Celular , Neoplasias de la Vulva/patología , Bélgica , Biomarcadores de Tumor/análisis , Biopsia , Carcinoma in Situ/química , Inhibidor p16 de la Quinasa Dependiente de Ciclina/análisis , Femenino , Humanos , Inmunohistoquímica , Países Bajos , Variaciones Dependientes del Observador , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Proteína p53 Supresora de Tumor/análisis , Neoplasias de la Vulva/química
7.
Hum Pathol ; 98: 81-88, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32142835

RESUMEN

Flat urothelial lesions with atypia may pose significant diagnostic challenges. Given frequent increased proliferation rates in florid reactive urothelial atypia and limited studies on the interpretation of p53 stains in the urothelium (following current standard guidelines for correlation with P53 mutation status), we sought to further study the discriminatory value of Ki-67 and p53 for florid reactive urothelial atypia versus urothelial carcinoma in situ (CIS). Bladder specimens diagnosed as reactive urothelial atypia (n = 40) and CIS (n = 40) were assessed by immunohistochemical staining with antibodies for Ki-67, p53, CD44, and CK20. Immunoreactivity was scored based on percent cells positive for Ki-67 and pattern of reactivity with p53 (aberrant: diffuse strong positive or negative; normal: patchy/wild type). CD44 and CK20 reactivity patterns served as adjunctive internal validation controls for reactive urothelial atypia and CIS, as previously described. In reactive urothelial atypia, Ki-67 ranged from 0% to 90% (mean, 34% ± 26) with 30 cases (75%) having >10%. In CIS, Ki-67 ranged from 5% to 95% (mean, 50% ± 25) with 17 cases (43%) having >50%. In all 40 cases (100%) of reactive urothelial atypia, p53 expression had a wild-type pattern. In CIS, aberrant p53 expression was identified in 15 cases (37%): 3 cases (7%) were p53 negative (i.e. null phenotype) and 12 cases (30%) showed strong and diffuse nuclear reactivity (in >85% of cells). The remaining 25 cases (63%) of CIS had a p53 wild-type pattern of expression. Cytoplasmic CK20 immunoreactivity in umbrella cells was seen in 34 cases (85%) of reactive urothelial atypia, and 6 cases (15%) were negative. In addition, 35 cases (88%) of reactive urothelial atypia demonstrated full-thickness CD44 expression, while 5 cases (12%) had expression confined to the basal/parabasal layers of the urothelium. Strong and diffuse CK20 positivity was present in 39 cases (98%) of CIS, and patchy positivity was detected in 1 case (2%). None of the CIS cases overexpressed CD44: 16 cases (40%) showed focal expression in the nonneoplastic basal cell layer; 24 cases (60%) demonstrated no staining. In summary, Ki-67 has poor discriminatory value for reactive urothelial atypia versus CIS and adds little to the classic CK20/CD44 immunophenotype. While p53 sensitivity for CIS is relatively low (30%) and interpretation as either wild type or negative may be challenging in a small subset of cases, strong and diffuse nuclear reactivity was 100% specific in the distinction from florid reactive urothelial atypia in this cohort.


Asunto(s)
Carcinoma in Situ/química , Proliferación Celular , Inmunohistoquímica , Antígeno Ki-67/análisis , Proteína p53 Supresora de Tumor/análisis , Neoplasias de la Vejiga Urinaria/química , Urotelio/química , Carcinoma in Situ/patología , Diagnóstico Diferencial , Humanos , Receptores de Hialuranos/análisis , Queratina-20/análisis , Valor Predictivo de las Pruebas , Neoplasias de la Vejiga Urinaria/patología , Urotelio/patología
8.
Int J Gynecol Pathol ; 39(6): 552-557, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31855954

RESUMEN

Although serous tubal intraepithelial carcinoma has been well described in the distal fallopian tube as precancers of pelvic high-grade serous carcinoma, endometrioid precancers have drawn less attention. Recently, endometrioid precursor lesions have been identified and reported to have a specific immunophenotype (PAX2-, ALDH1+, diffuse nuclear beta-catenin), as well as an association with both uterine and ovarian endometrioid carcinomas. These have been referred to as endometrioid (or type II) secretory cell outgrowths. A subset of endometrioid secretory cell outgrowths show architectural complexity resembling hyperplasia of the endometrium and have been referred to as endometrioid tubal intraepithelial neoplasia. We report 4 cases of endometrioid tubal intraepithelial neoplasia with clinical correlation and morphologic differential diagnosis.


Asunto(s)
Carcinoma in Situ/patología , Carcinoma Endometrioide/patología , Neoplasias de las Trompas Uterinas/patología , Lesiones Precancerosas/patología , Anciano , Carcinoma in Situ/química , Carcinoma Endometrioide/química , Diagnóstico Diferencial , Neoplasias Endometriales/química , Neoplasias Endometriales/patología , Trompas Uterinas/patología , Femenino , Humanos , Persona de Mediana Edad , Factor de Transcripción PAX2/análisis , Proteína p53 Supresora de Tumor/análisis , beta Catenina/análisis
9.
Am J Surg Pathol ; 43(12): 1638-1643, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31368912

RESUMEN

Although there are 5 well-described morphologic patterns of (nonglandular) urothelial carcinoma in situ (CIS), we have encountered a novel pattern of flat urothelial carcinoma with plasmacytoid features characterized by a triad of morphologic findings including abnormal architecture with cellular rounding, enlarged nuclei with eccentric nuclear localization, and dense globular eosinophilic cytoplasm. A total of 23 cases of plasmacytoid CIS (mean age: 74.1 y, range: 58 to 91 y) were collected and reviewed. We excluded cases in which the diagnostic biopsy had any of the following findings admixed in the same tissue biopsy sample as the plasmacytoid CIS: traditional patterns of CIS, noninvasive glandular CIS, papillary urothelial carcinoma, or invasive carcinoma. Immunostains for CK20, CD44, p53, and e-cadherin were performed on available blocks. History of prior urothelial neoplasia, prior treatment, and clinical follow-up were obtained from medical records and pathology re-review. Immunohistochemical analysis of plasmacytoid CIS showed diffuse/strong CK20 reactivity in 96% of cases (23/24), an abnormal p53 reactivity pattern (either overexpression or "null phenotype") in 37% of cases (7/19), absence of CD44 reactivity in the neoplastic cells in 63% of cases (15/24), and retained membranous e-cadherin expression in 100% of cases (18/18). Clinical follow-up (average follow-up time: 37.7 mo, range: 7 to 115 mo) showed recurrence/new occurrence in 52% of cases (12/23), including all 4 of the 23 patients who initially presented with de novo plasmacytoid CIS (ie, no prior or concomitant urothelial neoplasia). The histologic features, the immunophenotype, the association with other forms of urothelial neoplasia, and the risk of recurrence and progression in de novo lesions support that plasmacytoid CIS represents a novel pattern of flat urothelial carcinoma. These histologic features may be more subtle than in other more typical patterns of CIS and should be carefully distinguished from therapy-related/reactive changes.


Asunto(s)
Carcinoma in Situ/patología , Células Plasmáticas/patología , Neoplasias de la Vejiga Urinaria/patología , Urotelio/patología , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Carcinoma in Situ/química , Carcinoma in Situ/terapia , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Fenotipo , Células Plasmáticas/química , Factores de Riesgo , Resultado del Tratamiento , Neoplasias de la Vejiga Urinaria/química , Neoplasias de la Vejiga Urinaria/terapia , Urotelio/química
11.
Virchows Arch ; 475(1): 59-66, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31177317

RESUMEN

Sarcomatoid carcinomas recently came into the spotlight through genetic profiling studies and also as a distinct model of epithelial-mesenchymal transition. The literature on sarcomatoid carcinomas of gallbladder is limited. In this study, 656 gallbladder carcinomas (GBC) were reviewed. Eleven (1.7%) with a sarcomatoid component were identified and analyzed in comparison with ordinary GBC (O-GBC). Patients included 9 females and 2 males (F/M = 4.5 vs. 3.9) with a mean age-at-diagnosis of 71 (vs. 64). The median tumor size was 4.6 cm (vs. 2.5; P = 0.01). Nine patients (84%) presented with advanced stage (pT3/4) tumor (vs. 48%). An adenocarcinoma component constituting 1-75% of the tumor was present in nine, and eight had surface dysplasia/CIS; either in situ or invasive carcinoma was present in all cases. An intracholecystic papillary-tubular neoplasm was identified in one. Seven showed pleomorphic-sarcomatoid pattern, and four showed subtle/bland elongated spindle cells. Three had an angiosarcomatoid pattern. Two had heterologous elements. One showed few osteoclast-like giant cells, only adjacent to osteoid. Immunohistochemically, vimentin, was positive in six of six; P53 expression was > 60% in six of six, keratins in six of seven, and p63 in two of six. Actin, desmin, and S100 were negative. The median Ki67 index was 40%. In the follow-up, one died peri-operatively, eight died of disease within 3 to 8 months (vs. 26 months median survival for O-GBC), and two were alive at 9 and 15 months. The behavior overall was worse than ordinary adenocarcinomas in general but was not different when grade and stage were matched. In summary, sarcomatoid component is identified in < 2% of GBC. Unlike sarcomatoid carcinomas in the remainder of pancreatobiliary tract, these are seldom of the "osteoclastic" type and patients present with large/advanced stage tumors. Limited data suggests that these tumors are aggressive with rapid mortality unlike pancreatic osteoclastic ones which often have indolent behavior.


Asunto(s)
Adenocarcinoma/patología , Biomarcadores de Tumor/análisis , Carcinoma in Situ/patología , Neoplasias de la Vesícula Biliar/patología , Neoplasias Complejas y Mixtas/patología , Sarcoma/patología , Adenocarcinoma/química , Adenocarcinoma/mortalidad , Adenocarcinoma/cirugía , Anciano , Carcinoma in Situ/química , Carcinoma in Situ/mortalidad , Carcinoma in Situ/cirugía , Femenino , Neoplasias de la Vesícula Biliar/química , Neoplasias de la Vesícula Biliar/mortalidad , Neoplasias de la Vesícula Biliar/cirugía , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Neoplasias Complejas y Mixtas/química , Neoplasias Complejas y Mixtas/mortalidad , Neoplasias Complejas y Mixtas/cirugía , Sarcoma/química , Sarcoma/mortalidad , Sarcoma/cirugía , Factores de Tiempo , Resultado del Tratamiento
12.
Am J Surg Pathol ; 43(8): 1083-1091, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31107721

RESUMEN

Telomere alterations represent one of the major molecular changes in the development of human cancer. We have previously reported that telomere lengths in most serous tubal intraepithelial carcinomas (STIC) are shorter than they are in ovarian high-grade serous carcinomas (HGSC) or in normal-appearing fallopian tube epithelium from the same patients. However, it remains critical to determine if similar telomere alterations occur in TP53-mutated but histologically unremarkable "p53 signature" lesions, as well as incidental STICs without concurrent HGSC. In this study, we quantitatively measured telomere lengths by performing telomere-specific fluorescence in situ hybridization in conjunction with p53 immunolabeling in 15 p53 signatures and 30 incidental STICs without concurrent HGSC. We compared these new results with our previous data in paired STICs and concurrent HGSCs. We found that most p53 signatures (80%) and incidental STICs without HGSC (77%) exhibited significant telomere shortening compared with adjacent normal-appearing fallopian tube epithelium (P<0.01). Interestingly, however, p53 signatures and incidental STICs without HGSC displayed longer telomeres and less cell-to-cell telomere length heterogeneity than STICs associated with HGSC (P<0.001). These findings indicate that telomere shortening occurs in p53 signatures, the earliest precancer lesion. Moreover, incidental STICs without concurrent HGSC are indeed similar to p53 signatures as they have less telomere shortening and less cell-to-cell telomere length heterogeneity than STICs associated with HGSC.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma in Situ/genética , Neoplasias de las Trompas Uterinas/genética , Neoplasias Quísticas, Mucinosas y Serosas/genética , Acortamiento del Telómero , Telómero/genética , Transcriptoma , Proteína p53 Supresora de Tumor/genética , Biomarcadores de Tumor/análisis , Carcinoma in Situ/química , Carcinoma in Situ/patología , Carcinoma in Situ/cirugía , Neoplasias de las Trompas Uterinas/química , Neoplasias de las Trompas Uterinas/patología , Neoplasias de las Trompas Uterinas/cirugía , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Neoplasias Quísticas, Mucinosas y Serosas/química , Neoplasias Quísticas, Mucinosas y Serosas/patología , Neoplasias Quísticas, Mucinosas y Serosas/cirugía , Proteína p53 Supresora de Tumor/análisis
13.
Mod Pathol ; 32(8): 1095-1105, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30932019

RESUMEN

The switch from in situ to invasive tumor growth represents a crucial stage in the evolution of lung adenocarcinoma. However, the biological understanding of this shift is limited, and 'Noguchi Type C' tumors, being early lung adenocarcinomas with mixed in situ and invasive growth, represent those that are highly valuable in advancing our understanding of this process. All Noguchi Type C adenocarcinomas (n = 110) from the LATTICE-A cohort were reviewed and two patterns of in situ tumor growth were identified: those deemed likely to represent a true shift from precursor in situ to invasive disease ('Noguchi C1') and those in which the lepidic component appeared to represent outgrowth of the invasive tumor along existing airspaces ('Noguchi C2'). Overall Ki67 fraction was greater in C2 tumors and only C1 tumors showed significant increasing Ki67 from in situ to invasive disease. P53 positivity was acquired from in situ to invasive disease in C1 tumors but both components were positive in C2 tumors. Likewise, vimentin expression was increased from in situ to invasive tumor in C1 tumors only. Targeted next generation sequencing of 18 C1 tumors identified four mutations private to the invasive regions, including two in TP53, while 6 C2 tumors showed no private mutations. In the full LATTICe-A cohort, Ki67 fraction classified as either less than or greater than 10% within the in situ component of lung adenocarcinoma was identified as a strong predictor of patient outcome. This supports the proposition that tumors of all stages that have 'high grade' in situ components represent those with aggressive lepidic growth of the invasive clone. Overall these data support that the combined growth of Noguchi C tumors can represent two differing biological states and that 'Noguchi C1' tumors represent the genuine biological shift from in situ to invasive disease.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Carcinoma in Situ/patología , Proliferación Celular , Neoplasias Pulmonares/patología , Adenocarcinoma del Pulmón/química , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Carcinoma in Situ/química , Carcinoma in Situ/genética , Carcinoma in Situ/cirugía , Femenino , Humanos , Antígeno Ki-67/análisis , Neoplasias Pulmonares/química , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/cirugía , Masculino , Persona de Mediana Edad , Mutación , Clasificación del Tumor , Invasividad Neoplásica , Estadificación de Neoplasias , Estudios Retrospectivos , Proteína p53 Supresora de Tumor/análisis , Proteína p53 Supresora de Tumor/genética , Vimentina/análisis
14.
Am J Surg Pathol ; 42(11): 1556-1561, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30212393

RESUMEN

Invasive pancreatic ductal adenocarcinoma (PDAC) can infiltrate back into and spread along preexisting pancreatic ducts and ductules in a process known as cancerization of ducts (COD). Histologically COD can mimic high-grade pancreatic intraepithelial neoplasia (HG-PanIN). We reviewed pancreatic resections from 100 patients with PDAC for the presence or absence of ducts with histologic features of COD. Features supporting COD included adjacent histologically similar invasive PDAC and an abrupt transition between markedly atypical intraductal epithelium and normal duct epithelium or circumferential involvement of a duct. As the TP53 and SMAD4 genes are frequently targeted in invasive PDAC but not HG-PanIN, paired PDAC and histologically suspected COD lesions were immunolabeled with antibodies to the p53 and Smad4 proteins. Suspected COD was identified on hematoxylin and eosin sections in 89 (89%) of the cases. Immunolabeling for p53 and Smad4 was performed in 68 (76%) of 89 cases. p53 was interpretable in 55 cases and all 55 (100%) cases showed concordant labeling between COD and invasive PDAC. There was matched aberrant p53 immunolabeling in 37 (67%) cases including overexpression in 30 (55%) cases and lack of expression in 7 (13%) cases. Smad4 immunolabeling was interpretable in 61 cases and 59 (97%) cases showed concordant labeling between COD and invasive PDAC. Matched loss of Smad4 was seen in 28 (46%) cases. The immunolabeling of invasive PDAC and COD for p53 and Smad4 supports the high prevalence of COD observed on hematoxylin and eosin and highlights the utility of p53 and Smad4 immunolabeling in differentiating COD and HG-PanIN.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma in Situ/química , Carcinoma Ductal Pancreático/química , Inmunohistoquímica , Conductos Pancreáticos/química , Neoplasias Pancreáticas/química , Proteína Smad4/análisis , Proteína p53 Supresora de Tumor/análisis , Anciano , Anciano de 80 o más Años , Carcinoma in Situ/patología , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/cirugía , Bases de Datos Factuales , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Conductos Pancreáticos/patología , Conductos Pancreáticos/cirugía , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados
15.
Am J Surg Pathol ; 42(6): 828-835, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29505429

RESUMEN

Most human papillomavirus (HPV)-associated vulvar squamous cell carcinomas (VSCCs) originate from high-grade squamous intraepithelial lesions, also named usual type vulvar intraepithelial neoplasia. However, growing evidence suggests that morphologic studies have limitations in predicting HPV status in vulvar lesions. We aimed to evaluate adjacent intraepithelial lesions in a series of DNA HPV-positive VSCCs, focusing on unusual histologic patterns mimicking differentiated vulvar intraepithelial neoplasia (dVIN) or lichen sclerosus (LS). We identified 326 DNA HPV-positive VSCC with at least 1 cm of skin adjacent to the invasive tumor and analyzed HPV typing, HPV E6*I mRNA, and p16 immunohistochemistry in all cases. A careful histologic evaluation was conducted. A conclusive association with HPV was based on a positive p16 or HPV E6*I mRNA result or both in addition to the HPV DNA, whereas cases negative for both markers were classified as nonconclusively associated with HPV. One hundred twenty-one tumors (37.1%) had normal adjacent skin, 191 (58.6%) had only high-grade squamous intraepithelial lesions, also named usual type vulvar intraepithelial neoplasia, and unusual intraepithelial lesions were identified in 14 (4.3%) tumors. Seven cases showed dVIN-like features, 5 showed adjacent LS-like lesion, and in 2 cases dVIN-like and LS-like lesions were identified simultaneously. Six of them were conclusively associated with HPV (3 dVIN-like, 2 LS-like, 1 with combined dVIN/LS-like features). All 6 tumors were associated with HPV16 and were positive for both p16 and HPV mRNA, and p16 was also positive in the dVIN-like and LS-like lesions. In summary, a small subset of VSCCs conclusively associated with HPV may arise on intraepithelial lesions, mimicking precursors of HPV-independent VSCC.


Asunto(s)
Carcinoma in Situ/patología , Carcinoma de Células Escamosas/patología , Papillomavirus Humano 16/genética , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/patología , Proteínas Represoras/genética , Liquen Escleroso Vulvar/patología , Neoplasias de la Vulva/patología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Biopsia , Carcinoma in Situ/química , Carcinoma in Situ/virología , Carcinoma de Células Escamosas/química , Carcinoma de Células Escamosas/virología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/análisis , Diagnóstico Diferencial , Femenino , Pruebas de ADN del Papillomavirus Humano , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Clasificación del Tumor , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/virología , Valor Predictivo de las Pruebas , Neoplasias de la Vulva/química , Neoplasias de la Vulva/virología
16.
J Surg Res ; 223: 230-236, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29433879

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is among the most dismal of human malignancies. Neuropeptides have shown to be implicated in angiogenesis, tumor growth, and formation of distant metastases in various solid tumors. In the present study, we used a genetically engineered mouse model of pancreatic cancer to evaluate the impact of neuropeptide Y (NPY) and its receptors 1 (Y1) and 2 (Y2) in preneoplastic lesions and pancreatic cancer as a potential target with antiproliferative properties. In addition, human PDAC tissue was analyzed. MATERIALS AND METHODS: By interbreeding conditional LsL-Trp53R172H,LsL-KrasG12D and Pdx1-Cre strains, we obtained LsL-KrasG12D;LsL-Trp53R172H;Pdx1-Cre(KPC), LsL-KrasG12D;Pdx1-Cre(KP) and control mice (n = 8 each). Mice were then followed in a longitudinal study for 3 to 6 mo. Pancreata were analyzed in regard to pancreatic intraepithelial neoplasia (PanIN) lesions and invasive carcinoma. Corresponding sections were then assessed by immunohistochemistry and quantitative polymerase chain reaction for NPY, Y1 and Y2 expression in murine and human samples. RESULTS: NPY and Y1 expressions were detected in human and murine pancreatic samples, but expression levels were similar in neoplastic and non-neoplastic tissue. Y2 revealed a significant increase of expression in the transgenic mouse model in PanIN lesions and pancreatic cancer compared to control. This holds also true for human samples of pancreatic cancer. Immunohistochemistry of Y2 in murine and human samples of PanINs and pancreatic carcinoma revealed an increased expression in PanIN lesions and pancreatic cancer. CONCLUSIONS: Y2 is strongly overexpressed in pancreatic cancer and may modulate angiogenesis.


Asunto(s)
Carcinoma in Situ/química , Neuropéptido Y/análisis , Neoplasias Pancreáticas/química , Receptores de Neuropéptido Y/análisis , Animales , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Páncreas/química
17.
Am J Surg Pathol ; 42(4): 463-471, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29438174

RESUMEN

Progression of anal intraepithelial neoplasia (AIN) involves transition from productive to transforming human papillomavirus (HPV) infection. Grading aims to distinguish productive low-grade AIN from high-grade anal intraepithelial neoplasia (HGAIN) with risk of cancer. We describe immunohistochemical patterns in AIN adding a novel marker for initiation of the productive phase of the HPV life cycle (panHPVE4) to those for cell cycle activity (Ki-67) and transforming activity of HPVE7 gene (p16). We studied 67 anal biopsies for suspected anal neoplasia (17 normal, 15 AIN1, 20 AIN2, 15 AIN3) from 54 men who have sex with men at New York Presbyterian Hospital, USA. Two pathologists generated consensus AIN and immunogrades. Whole tissue and laser capture microdissection samples from multiple HPV-infected biopsies were tested for HPV with SPF10-PCR-DEIA-LiPA25, version 1. (Para)basal Ki-67 expression distinguished normal from AIN (≥lower-third Ki-67) with sensitivity 0.92 and specificity 1.0. Ki-67 did not distinguish grades of AIN. Null/patchy p16 versus diffuse ≥lower-third patterns discriminated HGAIN (sensitivity, 1.0; specificity, 0.84). There was marked heterogeneity in E4 expression within HGAIN. Most AIN2 (14/20) was E4 versus 0/15 AIN3 (sensitivity, 0.70; specificity 1.0). HPV was detected in 63 (94%) biopsies, with 49 (77.8%) high-risk HPV. HPV16 was the most frequent (13%). Multiple HPV genotypes were found in 15 (24%) biopsies and laser capture microdissection -polymerase chain reaction confirmed specific HPV types in E4 +/- AIN. Although Ki-67 discriminated AIN and p16 HGAIN, E4/p16 staining shows that most AIN2 is different from transformed AIN3 in showing both entry into productive HPV infection and transforming activity.


Asunto(s)
Neoplasias del Ano/química , Carcinoma in Situ/química , Inhibidor p16 de la Quinasa Dependiente de Ciclina/análisis , Pruebas de ADN del Papillomavirus Humano/métodos , Inmunohistoquímica , Antígeno Ki-67/análisis , Captura por Microdisección con Láser , Proteínas Oncogénicas Virales/análisis , Papillomaviridae/química , Infecciones por Papillomavirus/metabolismo , Reacción en Cadena de la Polimerasa , Neoplasias del Ano/patología , Neoplasias del Ano/virología , Biopsia , Carcinoma in Situ/patología , Carcinoma in Situ/virología , Transformación Celular Viral , ADN Viral/genética , Genotipo , Homosexualidad Masculina , Interacciones Huésped-Patógeno , Humanos , Masculino , Clasificación del Tumor , Ciudad de Nueva York , Papillomaviridae/genética , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Valor Predictivo de las Pruebas
18.
Mod Pathol ; 31(3): 406-417, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29148540

RESUMEN

Heterogeneous patterns of mutations and RNA expression have been well documented in invasive cancers. However, technological challenges have limited the ability to study heterogeneity of protein expression. This is particularly true for pre-invasive lesions such as ductal carcinoma in situ of the breast. Cell-level heterogeneity in ductal carcinoma in situ was analyzed in a single 5 µm tissue section using a multiplexed immunofluorescence analysis of 11 disease-related markers (EGFR, HER2, HER4, S6, pmTOR, CD44v6, SLC7A5 and CD10, CD4, CD8 and CD20, plus pan-cytokeratin, pan-cadherin, DAPI, and Na+K+ATPase for cell segmentation). Expression was quantified at cell level using a single-cell segmentation algorithm. K-means clustering was used to determine co-expression patterns of epithelial cell markers and immune markers. We document for the first time the presence of epithelial cell heterogeneity within ducts, between ducts and between patients with ductal carcinoma in situ. There was moderate heterogeneity in a distribution of eight clusters within each duct (average Shannon index 0.76; range 0-1.61). Furthermore, within each patient, the average Shannon index across all ducts ranged from 0.33 to 1.02 (s.d. 0.09-0.38). As the distribution of clusters within ducts was uneven, the analysis of eight ducts might be sufficient to represent all the clusters ie within- and between-duct heterogeneity. The pattern of epithelial cell clustering was associated with the presence and type of immune infiltrates, indicating a complex interaction between the epithelial tumor and immune system for each patient. This analysis also provides the first evidence that simultaneous analysis of both the epithelial and immune/stromal components might be necessary to understand the complex milieu in ductal carcinoma in situ lesions.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Proteínas de Neoplasias/metabolismo , Biomarcadores de Tumor/análisis , Neoplasias de la Mama/química , Carcinoma in Situ/química , Carcinoma in Situ/metabolismo , Carcinoma Ductal de Mama/química , Células Epiteliales/química , Células Epiteliales/patología , Femenino , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Proteínas de Neoplasias/análisis , Análisis de la Célula Individual
19.
Hum Pathol ; 67: 217-224, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28823571

RESUMEN

Von Meyenburg complex (VMC) is generally thought to be benign, although its preneoplastic potential for intrahepatic cholangiocarcinoma (iCC) has been a subject of contention. We retrospectively reviewed 86 hepatectomy specimens with a diagnosis of iCC. Morphologically, an association between iCC and VMC was appreciated in 35% of cases that illustrated a gradual neoplastic progression from benign VMC to dysplasia and then to iCC. Among them, 24 cases had VMC lined by epithelial cells with low-grade biliary dysplasia and 13 with high-grade biliary dysplasia. VMC-associated iCCs were smaller in size and well to moderately differentiated, with features of anastomosing glandular architecture, ductal carcinoma in situ-like growth pattern, peritumoral lymphocytic infiltrate, central fibrous scar, and complete pushing border. They often presented as T1 tumors. In contrast, non-VMC-associated iCCs were moderately to poorly differentiated with solid, cribriform or papillary growth patterns. They likely exhibited necrosis, perineural invasion, positive surgical margin, lymphovascular invasion, and high T stage. Additionally, Ki67 and p53 immunostains support the continuing neoplastic evolution from benign VMC to dysplasia and then to iCC. VMC could become neoplastic, serving as an in situ carcinoma lesion to transform to iCC. The underlying molecular alteration and clinical implication of this neoplastic transformation deserves further investigation.


Asunto(s)
Enfermedades de los Conductos Biliares/patología , Neoplasias de los Conductos Biliares/patología , Carcinoma in Situ/patología , Transformación Celular Neoplásica/patología , Colangiocarcinoma/patología , Lesiones Precancerosas/patología , Anciano , Anciano de 80 o más Años , Enfermedades de los Conductos Biliares/cirugía , Neoplasias de los Conductos Biliares/química , Neoplasias de los Conductos Biliares/cirugía , Biomarcadores de Tumor/análisis , Biopsia , Carcinoma in Situ/química , Carcinoma in Situ/cirugía , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/química , Colangiocarcinoma/química , Colangiocarcinoma/cirugía , Progresión de la Enfermedad , Femenino , Hepatectomía , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Lesiones Precancerosas/química , Lesiones Precancerosas/cirugía , Estudios Retrospectivos , Carga Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...