Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gut ; 71(5): 961-973, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-33849943

RESUMEN

OBJECTIVE: Recent studies have found aristaless-related homeobox gene (ARX)/pancreatic and duodenal homeobox 1 (PDX1), alpha-thalassemia/mental retardation X-linked (ATRX)/death domain-associated protein (DAXX) and alternative lengthening of telomeres (ALT) to be promising prognostic biomarkers for non-functional pancreatic neuroendocrine tumours (NF-PanNETs). However, they have not been comprehensively evaluated, especially among small NF-PanNETs (≤2.0 cm). Moreover, their status in neuroendocrine tumours (NETs) from other sites remains unknown. DESIGN: An international cohort of 1322 NETs was evaluated by immunolabelling for ARX/PDX1 and ATRX/DAXX, and telomere-specific fluorescence in situ hybridisation for ALT. This cohort included 561 primary NF-PanNETs, 107 NF-PanNET metastases and 654 primary, non-pancreatic non-functional NETs and NET metastases. The results were correlated with numerous clinicopathological features including relapse-free survival (RFS). RESULTS: ATRX/DAXX loss and ALT were associated with several adverse prognostic findings and distant metastasis/recurrence (p<0.001). The 5-year RFS rates for patients with ATRX/DAXX-negative and ALT-positive NF-PanNETs were 40% and 42% as compared with 85% and 86% for wild-type NF-PanNETs (p<0.001 and p<0.001). Shorter 5-year RFS rates for ≤2.0 cm NF-PanNETs patients were also seen with ATRX/DAXX loss (65% vs 92%, p=0.003) and ALT (60% vs 93%, p<0.001). By multivariate analysis, ATRX/DAXX and ALT status were independent prognostic factors for RFS. Conversely, classifying NF-PanNETs by ARX/PDX1 expression did not independently correlate with RFS. Except for 4% of pulmonary carcinoids, ATRX/DAXX loss and ALT were only identified in primary (25% and 29%) and NF-PanNET metastases (62% and 71%). CONCLUSIONS: ATRX/DAXX and ALT should be considered in the prognostic evaluation of NF-PanNETs including ≤2.0 cm tumours, and are highly specific for pancreatic origin among NET metastases of unknown primary.


Asunto(s)
Discapacidad Intelectual , Tumores Neuroendocrinos , Neoplasias Pancreáticas , Talasemia alfa , Proteínas Co-Represoras/genética , Genes Homeobox , Proteínas de Homeodominio , Humanos , Discapacidad Intelectual/genética , Chaperonas Moleculares/genética , Recurrencia Local de Neoplasia/genética , Tumores Neuroendocrinos/genética , Proteínas Nucleares/genética , Neoplasias Pancreáticas/patología , Telómero/genética , Telómero/patología , Factores de Transcripción/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Talasemia alfa/genética
2.
Ann Surg ; 275(6): e789-e795, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33201115

RESUMEN

OBJECTIVE: To evaluate the significance of UDD in IPMNs. BACKGROUND: The uncinate process of the pancreas has an independent ductal drainage system. International consensus guidelines of IPMNs still consider it as a branch-duct, even though it is the main drainage system for the uncinate process. METHODS: A retrospective review of all surgically treated IPMNs at our institution after 2008 was performed. Preoperative radiological studies were reviewed by an abdominal radiologist who was blinded to the pathological results. In addition to the Fukuoka criteria, presence of UDD was recorded. Using multivariate analysis, the pathological significance of UDD in predicting advanced neoplasia [high grade dysplasia or invasive carcinoma (HGD/ IC)] was determined. RESULTS: Two hundred sixty patients were identified (mean age at diagnosis was 68 years and 49% were females): 122 (47%) had HGD/IC. UDD was noted in 59 (23%), of which 36 (61%) had HGD/IC (P < 0.003). On multivariate analysis, UDD was an independent predictor of HGD/IC (odds ratio = 2.99, P < 0.04). Subgroup analysis on patients with IPMNs confined to the dorsal portion of the gland (n = 161), also demonstrated UDD to be a significant predictor of HGD/IC in those remote lesions (odds ratio: 4.41, P = 0.039). CONCLUSIONS: This is the largest study to evaluate the significance of UDD in IPMNs and shows it to be a high-risk feature. This association persisted for remote IPMNs limited to the dorsal pancreas, suggesting UDD may be associated with an aggressive phenotype even in remote IPMN lesions.


Asunto(s)
Adenocarcinoma Mucinoso , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Adenocarcinoma Mucinoso/diagnóstico por imagen , Adenocarcinoma Mucinoso/cirugía , Carcinoma Ductal Pancreático/diagnóstico por imagen , Carcinoma Ductal Pancreático/cirugía , Dilatación , Dilatación Patológica , Femenino , Humanos , Masculino , Páncreas/patología , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/cirugía , Estudios Retrospectivos
3.
Clin Gastroenterol Hepatol ; 20(4): 886-897, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33278573

RESUMEN

BACKGROUND & AIMS: The assessment of therapeutic response after neoadjuvant treatment and pancreatectomy for pancreatic ductal adenocarcinoma (PDAC) has been an ongoing challenge. Several limitations have been encountered when employing current grading systems for residual tumor. Considering endoscopic ultrasound (EUS) represents a sensitive imaging technique for PDAC, differences in tumor size between preoperative EUS and postoperative pathology after neoadjuvant therapy were hypothesized to represent an improved marker of treatment response. METHODS: For 340 treatment-naïve and 365 neoadjuvant-treated PDACs, EUS and pathologic findings were analyzed and correlated with patient overall survival (OS). A separate group of 200 neoadjuvant-treated PDACs served as a validation cohort for further analysis. RESULTS: Among treatment-naïve PDACs, there was a moderate concordance between EUS imaging and postoperative pathology for tumor size (r = 0.726, P < .001) and AJCC 8th edition T-stage (r = 0.586, P < .001). In the setting of neoadjuvant therapy, a decrease in T-stage correlated with improved 3-year OS rates (50% vs 31%, P < .001). Through recursive partitioning, a cutoff of ≥47% tumor size reduction was also found to be associated with improved OS (67% vs 32%, P < .001). Improved OS using a ≥47% threshold was validated using a separate cohort of neoadjuvant-treated PDACs (72% vs 36%, P < .001). By multivariate analysis, a reduction in tumor size by ≥47% was an independent prognostic factor for improved OS (P = .007). CONCLUSIONS: The difference in tumor size between preoperative EUS imaging and postoperative pathology among neoadjuvant-treated PDAC patients is an important prognostic indicator and may guide subsequent chemotherapeutic management.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/diagnóstico por imagen , Carcinoma Ductal Pancreático/cirugía , Endosonografía , Humanos , Terapia Neoadyuvante/métodos , Estadificación de Neoplasias , Pancreatectomía , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/cirugía , Pronóstico , Estudios Retrospectivos
4.
Gastroenterology ; 158(3): 573-582.e2, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31678302

RESUMEN

BACKGROUND & AIMS: Intraductal oncocytic papillary neoplasms (IOPNs) of the pancreas and bile duct contain epithelial cells with numerous, large mitochondria and are cystic precursors to pancreatic ductal adenocarcinoma (PDAC) and cholangiocarcinoma (CCA), respectively. However, IOPNs do not have the genomic alterations found in other pancreatobiliary neoplasms. In fact, no recurrent genomic alterations have been described in IOPNs. PDACs without activating mutations in KRAS contain gene rearrangements, so we investigated whether IOPNs have recurrent fusions in genes. METHODS: We analyzed 20 resected pancreatic IOPNs and 3 resected biliary IOPNs using a broad RNA-based targeted sequencing panel to detect cancer-related fusion genes. Four invasive PDACs and 2 intrahepatic CCAs from the same patients as the IOPNs, were also available for analysis. Samples of pancreatic cyst fluid (n = 5, collected before surgery) and bile duct brushings (n = 2) were analyzed for translocations. For comparison, we analyzed pancreatobiliary lesions from 126 patients without IOPN (controls). RESULTS: All IOPNs evaluated were found to have recurring fusions of ATP1B1-PRKACB (n = 13), DNAJB1-PRKACA (n = 6), or ATP1B1-PRKACA (n = 4). These fusions also were found in corresponding invasive PDACs and intrahepatic CCAs, as well as in matched pancreatic cyst fluid and bile duct brushings. These gene rearrangements were absent from all 126 control pancreatobiliary lesions. CONCLUSIONS: We identified fusions in PRKACA and PRKACB genes in pancreatic and biliary IOPNs, as well as in PDACs and pancreatic cyst fluid and bile duct cells from the same patients. We did not identify these gene fusions in 126 control pancreatobiliary lesions. These fusions might be used to identify patients at risk for IOPNs and their associated invasive carcinomas.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Carcinoma Ductal Pancreático/genética , Colangiocarcinoma/genética , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/genética , Neoplasias Intraductales Pancreáticas/genética , Neoplasias Pancreáticas/genética , Adulto , Anciano , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos , Femenino , Fusión Génica , Reordenamiento Génico , Proteínas del Choque Térmico HSP40/genética , Humanos , Masculino , Persona de Mediana Edad , Quiste Pancreático/genética , Neoplasias Intraductales Pancreáticas/patología , Neoplasias Pancreáticas/patología , ATPasa Intercambiadora de Sodio-Potasio/genética
5.
Gut ; 69(1): 52-61, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30971436

RESUMEN

OBJECTIVE: Despite improvements in imaging, serum CA19-9 and pathological evaluation, differentiating between benign and malignant bile duct strictures remains a diagnostic conundrum. Recent developments in next-generation sequencing (NGS) have opened new opportunities for early detection and management of cancers but, to date, have not been rigorously applied to biliary specimens. DESIGN: We prospectively evaluated a 28-gene NGS panel (BiliSeq) using endoscopic retrograde cholangiopancreatography-obtained biliary specimens from patients with bile duct strictures. The diagnostic performance of serum CA19-9, pathological evaluation and BiliSeq was assessed on 252 patients (57 trainings and 195 validations) with 346 biliary specimens. RESULTS: The sensitivity and specificity of BiliSeq for malignant strictures was 73% and 100%, respectively. In comparison, an elevated serum CA19-9 and pathological evaluation had sensitivities of 76% and 48%, and specificities of 69% and 99%, respectively. The combination of BiliSeq and pathological evaluation increased the sensitivity to 83% and maintained a specificity of 99%. BiliSeq improved the sensitivity of pathological evaluation for malignancy from 35% to 77% for biliary brushings and from 52% to 83% for biliary biopsies. Among patients with primary sclerosing cholangitis (PSC), BiliSeq had an 83% sensitivity as compared with pathological evaluation with an 8% sensitivity. Therapeutically relevant genomic alterations were identified in 20 (8%) patients. Two patients with ERBB2-amplified cholangiocarcinoma received a trastuzumab-based regimen and had measurable clinicoradiographic response. CONCLUSIONS: The combination of BiliSeq and pathological evaluation of biliary specimens increased the detection of malignant strictures, particularly in patients with PSC. Additionally, BiliSeq identified alterations that may stratify patients for specific anticancer therapies.


Asunto(s)
Colangiopancreatografia Retrógrada Endoscópica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de los Conductos Biliares/diagnóstico , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Enfermedades de las Vías Biliares/diagnóstico , Enfermedades de las Vías Biliares/genética , Enfermedades de las Vías Biliares/patología , Biomarcadores de Tumor/sangre , Antígeno CA-19-9/sangre , Constricción Patológica/diagnóstico , Constricción Patológica/genética , Diagnóstico Diferencial , Femenino , Humanos , Cirrosis Hepática Biliar/diagnóstico , Cirrosis Hepática Biliar/genética , Cirrosis Hepática Biliar/patología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Sensibilidad y Especificidad , Manejo de Especímenes/métodos , Adulto Joven
6.
Histopathology ; 77(3): 481-491, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32431062

RESUMEN

AIMS: Abnormal p53 protein expression detected by immunohistochemistry (IHC) in Barrett's oesophagus (BO) is reported to be a prognostic biomarker for progression to high-grade dysplasia (HGD) or oesophageal adenocarcinoma (OAC). We evaluated our use of p53 IHC for patients with BO under surveillance from 2010 to 2016 in a single academic institution. METHODS AND RESULTS: We identified 78 patients under surveillance for BO who had biopsies evaluated for abnormal p53 expression in conjunction with routine histology and 892 patients who had histological evaluation alone. All available p53 IHC slides were rescored as wild-type or abnormal. We evaluated the risk of subsequent diagnosis with HGD and OAC. p53-tested patients were significantly more likely to be diagnosed with indefinite dysplasia (IND) or low-grade dysplasia (LGD), compared to patients who were not tested (79.5 versus 10.8%, P = 7.4 × 10-40 ). Almost half (46.9%) of patients with abnormal p53 expression were diagnosed with HGD or OAC within 5 years, compared to 5.9% with wild-type p53, and 7.6% of patients not tested (P = 2.6 × 10-18 ). However, this difference was heavily influenced by other risk factors, including dysplasia grade, in multivariate analyses. In the subgroup of patients diagnosed with IND (n = 109), abnormal p53 expression was associated with a fourfold increase (1.2-13.3, P = 0.023) in risk of HGD/OAC relative to untested patients diagnosed with IND, independent of other risk factors. CONCLUSION: In patients under surveillance for BO in a single academic institution, we found evidence that selective use of p53 IHC in conjunction with routine histology modestly improved risk stratification by identifying patients with IND at higher risk of a subsequent diagnosis of HGD or OAC.


Asunto(s)
Esófago de Barrett/patología , Biomarcadores de Tumor/análisis , Lesiones Precancerosas/patología , Proteína p53 Supresora de Tumor/biosíntesis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Adulto Joven
7.
Gastroenterology ; 154(8): 2060-2063.e8, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29486199

RESUMEN

Despite prognostic grading and staging systems, it is a challenge to predict outcomes for patients with pancreatic neuroendocrine tumors (PanNETs). Sequencing studies of PanNETs have identified alterations in death domain-associated protein (DAXX) and alpha-thalassemia/mental retardation X-linked chromatin remodeler (ATRX). In tumors, mutations in DAXX or ATRX and corresponding loss of protein expression correlate with shorter times of disease-free survival and disease-specific survival of patients. However, DAXX or ATRX proteins were lost in only 50% of distant metastases analyzed. We performed whole-exome sequencing analyses of 20 distant metastases from 20 patients with a single nonsyndrome, nonfunctional PanNET. We found distant metastases contained alterations in multiple endocrine neoplasia type 1 (MEN1) (n = 8), ATRX (n = 5), DAXX (n = 5), TSC2 (n = 3), and DEP domain containing 5 (DEPDC5) (n = 3). We found copy number loss of cyclin dependent kinase inhibitor 2A (CDKN2A) in 15 metastases (75%) and alterations in genes that regulate chromatin remodeling, including set domain containing 2 (SETD2) (n = 4), AT-rich interaction domain 1A (ARID1A) (n = 2), chromodomain helicase DNA binding protein 8 (CHD8) (n = 2), and DNA methyl transferase 1 (DNMT1) (n = 2). In a separate analysis of 347 primary PanNETs, we found loss or deletion of DAXX and ATRX, disruption of SETD2 function (based on loss of H3 lysine 36 trimethylation), loss of ARID1A expression or deletions in CDKN2A in 81% of primary PanNETs with distant metastases. Among patients with loss or deletion of at least 1 of these proteins or genes, 39% survived disease-free for 5 years and 44% had disease-specific survival times of 10 years. Among patients without any of these alterations, 98% survived disease-free for 5 years and 95% had disease-specific survival times of 10 years. Therefore, primary PanNETs with loss of DAXX, ATRX, H3 lysine 36 trimethylation, ARID1A, and/or CDKN2A associate with shorter survival times of patients. Our findings indicate that alterations in chromatin-remodeling genes and CDKN2A contribute to metastasis of PanNETs.


Asunto(s)
Biomarcadores de Tumor/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Tumores Neuroendocrinos/genética , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Adulto , Anciano , Anciano de 80 o más Años , Ensamble y Desensamble de Cromatina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Variaciones en el Número de Copia de ADN , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mutación , Tumores Neuroendocrinos/mortalidad , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/cirugía , Pancreatectomía , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Pronóstico , Secuenciación del Exoma
8.
Gut ; 67(12): 2131-2141, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-28970292

RESUMEN

OBJECTIVE: DNA-based testing of pancreatic cyst fluid (PCF) is a useful adjunct to the evaluation of pancreatic cysts (PCs). Mutations in KRAS/GNAS are highly specific for intraductal papillary mucinous neoplasms (IPMNs) and mucinous cystic neoplasms (MCNs), while TP53/PIK3CA/PTEN alterations are associated with advanced neoplasia. A prospective study was performed to evaluate preoperative PCF DNA testing. DESIGN: Over 43-months, 626 PCF specimens from 595 patients were obtained by endoscopic ultrasound (EUS)-fine needle aspiration and assessed by targeted next-generation sequencing (NGS). Molecular results were correlated with EUS findings, ancillary studies and follow-up. A separate cohort of 159 PCF specimens was also evaluated for KRAS/GNAS mutations by Sanger sequencing. RESULTS: KRAS/GNAS mutations were identified in 308 (49%) PCs, while alterations in TP53/PIK3CA/PTEN were present in 35 (6%) cases. Based on 102 (17%) patients with surgical follow-up, KRAS/GNAS mutations were detected in 56 (100%) IPMNs and 3 (30%) MCNs, and associated with 89% sensitivity and 100% specificity for a mucinous PC. In comparison, KRAS/GNAS mutations by Sanger sequencing had a 65% sensitivity and 100% specificity. By NGS, the combination of KRAS/GNAS mutations and alterations in TP53/PIK3CA/PTEN had an 89% sensitivity and 100% specificity for advanced neoplasia. Ductal dilatation, a mural nodule and malignant cytopathology had lower sensitivities (42%, 32% and 32%, respectively) and specificities (74%, 94% and 98%, respectively). CONCLUSIONS: In contrast to Sanger sequencing, preoperative NGS of PCF for KRAS/GNAS mutations is highly sensitive for IPMNs and specific for mucinous PCs. In addition, the combination of TP53/PIK3CA/PTEN alterations is a useful preoperative marker for advanced neoplasia.


Asunto(s)
Biomarcadores de Tumor/genética , Líquido Quístico/química , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Quiste Pancreático/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/cirugía , Cromograninas/genética , ADN de Neoplasias/genética , Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico , Femenino , Estudios de Seguimiento , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación , Proteínas de Neoplasias/genética , Neoplasias Quísticas, Mucinosas y Serosas/diagnóstico , Neoplasias Quísticas, Mucinosas y Serosas/genética , Neoplasias Quísticas, Mucinosas y Serosas/cirugía , Quiste Pancreático/genética , Quiste Pancreático/patología , Quiste Pancreático/cirugía , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Cuidados Preoperatorios , Estudios Prospectivos , Proteínas Proto-Oncogénicas p21(ras)/genética , Sensibilidad y Especificidad , Adulto Joven
9.
Gastrointest Endosc ; 83(6): 1107-1117.e2, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26709110

RESUMEN

BACKGROUND AND AIMS: The American Gastroenterological Association (AGA) recently reported evidence-based guidelines for the management of asymptomatic neoplastic pancreatic cysts. These guidelines advocate a higher threshold for surgical resection than prior guidelines and imaging surveillance for a considerable number of patients with pancreatic cysts. The aims of this study were to assess the accuracy of the AGA guidelines in detecting advanced neoplasia and present an alternative approach to pancreatic cysts. METHODS: The study population consisted of 225 patients who underwent EUS-guided FNA for pancreatic cysts between January 2014 and May 2015. For each patient, clinical findings, EUS features, cytopathology results, carcinoembryonic antigen analysis, and molecular testing of pancreatic cyst fluid were reviewed. Molecular testing included the assessment of hotspot mutations and deletions for KRAS, GNAS, VHL, TP53, PIK3CA, and PTEN. RESULTS: Diagnostic pathology results were available for 41 patients (18%), with 13 (6%) harboring advanced neoplasia. Among these cases, the AGA guidelines identified advanced neoplasia with 62% sensitivity, 79% specificity, 57% positive predictive value, and 82% negative predictive value. Moreover, the AGA guidelines missed 45% of intraductal papillary mucinous neoplasms with adenocarcinoma or high-grade dysplasia. For cases without confirmatory pathology, 27 of 184 patients (15%) with serous cystadenomas (SCAs) based on EUS findings and/or VHL alterations would continue magnetic resonance imaging (MRI) surveillance. In comparison, a novel algorithmic pathway using molecular testing of pancreatic cyst fluid detected advanced neoplasias with 100% sensitivity, 90% specificity, 79% positive predictive value, and 100% negative predictive value. CONCLUSIONS: The AGA guidelines were inaccurate in detecting pancreatic cysts with advanced neoplasia. Furthermore, because the AGA guidelines manage all neoplastic cysts similarly, patients with SCAs will continue to undergo unnecessary MRI surveillance. The results of an alternative approach with integrative molecular testing are encouraging but require further validation.


Asunto(s)
Carcinoma Ductal Pancreático/diagnóstico , Neoplasias Quísticas, Mucinosas y Serosas/diagnóstico , Quiste Pancreático/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Guías de Práctica Clínica como Asunto , Adenocarcinoma/sangre , Adenocarcinoma/diagnóstico , Adenocarcinoma/genética , Adenocarcinoma/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antígeno Carcinoembrionario/sangre , Carcinoma Ductal Pancreático/sangre , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Cromograninas/genética , Fosfatidilinositol 3-Quinasa Clase I , Líquido Quístico , Cistadenoma Seroso/sangre , Cistadenoma Seroso/diagnóstico , Cistadenoma Seroso/genética , Cistadenoma Seroso/patología , Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico , Endosonografía , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Técnicas de Diagnóstico Molecular , Neoplasias Quísticas, Mucinosas y Serosas/sangre , Neoplasias Quísticas, Mucinosas y Serosas/genética , Neoplasias Quísticas, Mucinosas y Serosas/patología , Fosfohidrolasa PTEN/genética , Quiste Pancreático/sangre , Quiste Pancreático/genética , Quiste Pancreático/patología , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Estudios Retrospectivos , Sensibilidad y Especificidad , Proteína p53 Supresora de Tumor/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Adulto Joven
10.
J Infect Dis ; 211(10): 1560-5, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25231015

RESUMEN

Human polyomavirus 7 (HPyV7) is one of 11 HPyVs recently discovered through genomic sequencing technologies. Two lung transplant recipients receiving immunosuppressive therapy developed pruritic, brown plaques on the trunk and extremities showing a distinctive epidermal hyperplasia with virus-laden keratinocytes containing densely packed 36-45-nm icosahedral capsids. Rolling circle amplification and gradient centrifugation testing were positive for encapsidated HPyV7 DNA in skin and peripheral blood specimens from both patients, and HPyV7 early and capsid proteins were abundantly expressed in affected tissues. We describe for the first time that HPyV7 is associated with novel pathogenicity in some immunosuppressed individuals.


Asunto(s)
Infecciones por Polyomavirus/patología , Infecciones por Polyomavirus/virología , Poliomavirus/aislamiento & purificación , Receptores de Trasplantes , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología , Anciano , Sangre/virología , Exantema/patología , Exantema/virología , Histocitoquímica , Humanos , Huésped Inmunocomprometido , Inmunohistoquímica , Masculino , Microscopía Electrónica de Transmisión , Piel/patología , Piel/virología , Viremia
12.
Pancreas ; 53(4): e350-e356, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38518061

RESUMEN

BACKGROUND: The International Cancer of the Pancreas Screening Consortium recommended annual imaging for individuals at increased risk for developing a pancreatic ductal adenocarcinoma (PDAC) who did not have concerning pancreatic findings or a cyst <3 cm without worrisome features. We aimed to determine if 3-cm cyst size accurately predicted advanced precursor lesions in high-risk individuals undergoing surveillance. METHODS: Imaging for high-risk individuals (HRIs) undergoing PDAC surveillance from 2007 to 2021 was reviewed and pancreatic abnormalities were recorded including dominant cyst size and number of cysts. Subjects were excluded if they had the following: (1) no follow-up imaging after baseline, (2) solid lesion at baseline, or (3) development of solid lesion without having cyst on prior imaging. RESULTS: Five of the 77 HRIs found to have a cystic lesion on surveillance developed a PDAC: 3 with cystic lesion >1 cm as compared with only 2 of 67 HRIs with cystic lesions <1 cm (P < 0.05). None of these cysts developed worrisome findings and 4/5 PDACs did not arise from visualized cystic precursor lesion. CONCLUSIONS: Patients with a cyst ≥1 cm were at increased risk for developing PDAC compared with patients with cyst <1 cm. Pancreatic ductal adenocarcinoma usually did not arise from a recognized cystic lesion.


Asunto(s)
Carcinoma Ductal Pancreático , Quiste Pancreático , Neoplasias Pancreáticas , Humanos , Imagen por Resonancia Magnética/métodos , Neoplasias Pancreáticas/patología , Quiste Pancreático/diagnóstico , Páncreas/patología , Carcinoma Ductal Pancreático/patología , Estudios Retrospectivos
13.
Clin Gastroenterol Hepatol ; 11(4): 425-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23267865

RESUMEN

BACKGROUND & AIMS: Endoscopic ultrasound (EUS) with fine-needle aspiration is routinely used to evaluate pancreatic cysts. We investigated the association between results from DNA analysis of cyst fluid and patient outcomes. METHODS: In a retrospective analysis, we collected data from 113 patients with pancreatic cysts who underwent EUS with fine-needle aspiration at a tertiary care center from June 2004 to June 2007. Detailed follow-up data were obtained through October 2010 (mean, 47 months). Pancreatic cysts were categorized as nonbenign or benign on the basis of pathology analysis of surgical samples and patients' outcomes. We compared the patient characteristics, presenting symptoms, EUS imaging characteristics, and results from analysis of cyst fluid, including cytology results, levels of carcinoembryonic antigen, and DNA sequencing results. RESULTS: Fifty-one patients underwent pancreatic surgery (10 had malignant, 18 had mucinous, and 16 had benign cysts), 63 patients were followed long-term, and 13 patients died of pancreatic cancer. On the basis of multivariate regression analysis, the presence of cyst solid component, patient symptoms, cyst size >3 cm, and detection of KRAS mutations at codons 12 and 13 in cyst fluid were independently associated with a nonbenign course. CONCLUSIONS: KRAS mutations, detected in pancreatic cyst fluid, are associated with mucinous cysts and progression and development of malignancy and should be considered in assessing pancreatic cysts. The presence of a cyst solid component, patient symptoms, and cyst size greater than 3 cm were additional high-risk factors for a malignant course of disease.


Asunto(s)
Quiste Pancreático/genética , Quiste Pancreático/patología , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Anciano , Anciano de 80 o más Años , Biopsia con Aguja Fina , Líquido Quístico/química , ADN/química , ADN/genética , ADN/aislamiento & purificación , Endosonografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas p21(ras) , Estudios Retrospectivos , Análisis de Secuencia de ADN , Centros de Atención Terciaria , Resultado del Tratamiento
14.
Mod Pathol ; 26(11): 1478-87, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23743931

RESUMEN

With improvements in abdominal imaging, detection of incidental pancreatic cysts are becoming increasingly common. Analysis of pancreatic cyst fluid from fine-needle aspiration is particularly important in identifying intraductal papillary mucinous neoplasms (IPMNs) and mucinous cystic neoplasms (MCNs), which have significant implications in clinical intervention and follow-up. Previous controlled studies have shown that KRAS mutations in cyst fluid are highly specific for mucinous differentiation in pancreatic cysts; however, this has not been examined in the clinical setting. Over a 6-year study period, 618 pancreatic cyst fluids obtained by fine-needle aspiration at the time of endoscopic ultrasound were tested for KRAS mutations as part of routine evaluation for a cystic neoplasm. Of the 618 specimens, 603 (98%) from 546 patients were satisfactory for molecular analysis. Patients ranged in age from 17 to 90 years (mean, 63.9 years) and were predominantly female (68%). Pancreatic cysts were relatively evenly distributed throughout the pancreas and ranged in size from 0.6 to 11.0 cm (mean, 2.3 cm). Mutations in KRAS were detected in 232 of 603 (38%) aspirates. Although sufficient for molecular analysis, 320 of 603 (53%) specimens were either less than optimal (38%) or unsatisfactory (15%) for cytopathologic diagnosis. Surgical follow-up information was available for 142 (26%) patients and consisted of 53 KRAS-mutated and 89 KRAS-wild-type cysts. Overall, KRAS mutations had a specificity of 100%, but a sensitivity of 54% for mucinous differentiation. When stratified by cyst type, KRAS had a sensitivity of 67% and 14% for IPMNs and MCNs, respectively. In summary, KRAS mutations were highly specific for mucinous differentiation, but were inadequate in identifying MCNs. Future molecular studies and the combination of other fluid markers are required to improve the detection and classification of pancreatic mucinous neoplasms by endoscopic ultrasound fine-needle aspiration.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Análisis Mutacional de ADN , Pruebas Genéticas/métodos , Mutación , Neoplasias Quísticas, Mucinosas y Serosas/genética , Quiste Pancreático/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/patología , Diferenciación Celular , Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Quísticas, Mucinosas y Serosas/patología , Quiste Pancreático/patología , Neoplasias Pancreáticas/patología , Fenotipo , Valor Predictivo de las Pruebas , Pronóstico , Proteínas Proto-Oncogénicas p21(ras) , Adulto Joven
15.
Gastrointest Endosc ; 77(6): 872-6, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23472998

RESUMEN

BACKGROUND: EMR and ablation are increasingly being used alone or in combination for treatment of Barrett's neoplasia. Given a very low rate of lymph node metastasis, endotherapy has become an accepted treatment option for T1a esophageal adenocarcinoma (EAC) with low-risk features. OBJECTIVE: To report our experience of endoscopic management of T1a EAC in a large, tertiary-care center. DESIGN: Retrospective review. SETTING: Tertiary-care referral center. PATIENTS: Patients treated endoscopically for low-risk T1a EAC at our center. INTERVENTION: EMR and endoscopic ablation. MAIN OUTCOME MEASUREMENTS: Death related to esophageal cancer, remission of adenocarcinoma, dysplasia, and intestinal metaplasia. RESULTS: A total of 54 patients underwent endotherapy for low-risk T1a EAC from 2006 to 2012. Mean (± SD) follow-up was 23 (± 16) months, mean (± SD) size of resected adenocarcinoma was 7.1 (± 4.3) mm, and mean (± SD) Barrett's esophagus length was 4.5 (± 3.9) cm. Band-assisted, cap-assisted, and lift and cut EMR were performed in 85%, 11%, and 4% of patients, respectively; 81% underwent additional ablative therapy (radiofrequency ablation 95%, cryotherapy 9%, photodynamic therapy 2%). Complete remission from cancer was achieved in 96%, complete remission from dysplasia in 87%, and complete remission from intestinal metaplasia in 59%. The overall survival was 89%; there were no deaths related to esophageal cancer. LIMITATIONS: Retrospective study. CONCLUSION: Endotherapy for T1a EAC was safe and effective in our American cohort. Endotherapy should be considered primary therapy for appropriate patients with low-risk lesions. Complete Barrett's esophagus eradication after EMR is important to reduce the development of metachronous lesions.


Asunto(s)
Adenocarcinoma/cirugía , Neoplasias Esofágicas/cirugía , Membrana Mucosa/cirugía , Adenocarcinoma/patología , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Neoplasias Esofágicas/patología , Esofagoscopía/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Membrana Mucosa/patología , Estudios Retrospectivos , Resultado del Tratamiento , Estados Unidos
16.
Pancreatology ; 11(5): 482-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21997479

RESUMEN

BACKGROUND/AIMS: Autoimmune pancreatitis (AIP) may mimic pancreatic cancer (PC). The detection of DNA mutations in endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) material may improve discrimination between AIP and PC and is the context for this study. METHODS: In a retrospective study, archived EUS-FNA material from patients with AIP and PC at two centers was analyzed for KRAS mutations and loss-of-heterozygosity analysis involving 18 microsatellite markers. KRAS status and the fractional allelic loss (number of affected microsatellites divided by informative ones) were compared for AIP and PC. RESULTS: Thirty-two patients with 33 samples were studied. There were 16 patients with AIP (17 samples) and 16 patients with PC. DNA amplification failed in 7 samples. Of 25 patients (26 samples), 14 had AIP (7 male, age 57 ± 17 years; mean ± SD) and 11 had PC (7 male, age 65 ± 14 years; mean ± SD). Cytology results for AIP were inflammatory = 3, inconclusive = 10, suspicious for malignancy = 2 and for PC were malignant = 5, suspicious for malignancy = 4 and inconclusive = 2, respectively. KRAS mutation was detected in none of the AIP cases and 10/11 PC cases (91%, Pearson χ(2) = 22.16, p < 0.001) or 10/16 PC cases (63%) accounting for PC cases with failed DNA amplification. Mean (±SD) fractional allelic loss for the AIP cases (0.16 ± 0.15) was not significantly different from the PC cases (0.26 ± 0.19). CONCLUSIONS: A KRAS mutation in EUS/FNA material from a pancreatic mass is associated with malignancy and may help discriminate from benign conditions such as AIP.


Asunto(s)
Enfermedades Autoinmunes/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Pancreatitis/diagnóstico , Adulto , Anciano , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Biopsia con Aguja Fina/métodos , Análisis Mutacional de ADN , Endosonografía , Femenino , Genes ras/genética , Humanos , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Pancreatitis/genética , Pancreatitis/patología , Estudios Retrospectivos
17.
Pancreatology ; 10(4): 429-33, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20720444

RESUMEN

BACKGROUND/AIMS: Preoperative distinction between pancreatic cancer (PC) and extrahepatic cholangiocarcinoma (CC) is desirable due to diverging management options, and to optimize enrollment into neoadjuvant trials. METHODS: A single-center retrospective study of patients with PC or CC was undertaken. Four blinded pathologists reviewed all cases and reached a consensus diagnosis (PC or CC). Microdissection-based multiple microsatellite loss analysis and direct sequencing of K-ras oncogene was performed and compared for PC and CC. RESULTS: Of 33 cases studied (17 males; 16 PC, 17 CC; 10 with primary sclerosing cholangitis), a K-ras mutation was present in 14/16 (87.5%) PC and 1/17 (5.9%) CC cases (p < 0.001), sensitivity and specificity were 87.5 and 94%, respectively. The mean fractional mutational rate was higher in PC (0.51; 95% CI 0.45-0.58) compared to CC (0.34; 95% CI 0.28-0.39, p < 0.001). CONCLUSIONS: The presence of a K-ras mutation in cytology specimens distinguishes PC from CC in this study. and IAP.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos/patología , Carcinoma Ductal Pancreático/genética , Colangiocarcinoma/genética , Análisis Mutacional de ADN , Neoplasias Pancreáticas/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de los Conductos Biliares/diagnóstico , Carcinoma Ductal Pancreático/diagnóstico , Colangiocarcinoma/diagnóstico , Colangitis Esclerosante/genética , Colangitis Esclerosante/patología , ADN de Neoplasias/análisis , Femenino , Genes ras/genética , Humanos , Masculino , Microdisección , Inestabilidad de Microsatélites , Persona de Mediana Edad , Mutación , Neoplasias Pancreáticas/diagnóstico , Valor Predictivo de las Pruebas , Estudios Retrospectivos , Sensibilidad y Especificidad
18.
Gastrointest Endosc ; 71(7): 1178-84, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20362284

RESUMEN

BACKGROUND: Stereotactic body radiotherapy (SBRT) has been approved for the treatment of locally advanced pancreatic cancer. Placement of gold fiducials is required for real-time tracking and delivery of a high-dose therapeutic beam of radiation to the tumor. Traditionally, fiducials have been placed either intraoperatively or percutaneously. Recently, EUS-guided fiducial placement has been reported, but the safety and feasibility of this approach is not well defined. OBJECTIVE: The aim of this study was to determine the safety, feasibility, and limitations of EUS-guided placement of 0.8 x 5.0 mm fiducials via a 19-gauge needle for locally advanced and recurrent pancreatic cancer. DESIGN: Prospective study of patients with either locally advanced or recurrent pancreatic cancer referred for EUS-guided fiducial placement for SBRT at our institution over a 3-year period. SETTING: Tertiary referral center conducting >1800 EUS procedures annually. MAIN OUTCOME MEASUREMENTS: Primary outcome measurements included success, complications, and technical limitations of EUS-guided fiducial placement in pancreatic cancer. In addition, the percentage of patients successfully completing SBRT after EUS-guided fiducial placement was determined. RESULTS: A total of 51 patients (mean age 73 years; 57% male) with locally advanced (n = 36) and recurrent (n = 15) pancreatic cancer were referred for EUS-guided fiducial placement. Fiducials were successfully placed in 46 patients (90%), with technical failures occurring in 4 patients (8%) with recurrent cancer after pancreaticoduodenectomy. In 3 patients (7%), the fiducials spontaneously migrated from the original site of injection, thereby requiring a second EUS procedure for placement of additional fiducials. Of the 46 patients with fiducials placed under EUS guidance, 42 patients (91%) successfully completed SBRT. Two patients experienced disease progression before SBRT, 1 patient was lost to follow-up, and 1 patient experienced a complication at ERCP that precluded further therapy. Only 1 complication (2%), of mild pancreatitis, occurred in a patient undergoing simultaneous placement of fiducials and celiac plexus neurolysis for intractable abdominal pain. LIMITATIONS: Single-center experience and lack of a formal follow-up protocol to assess for complications. CONCLUSION: EUS-guided fiducial placement for SBRT in locally advanced and recurrent pancreatic cancer is safe and feasible. Successful placement was achieved in 90% of patients, with a low complication rate (2%). Furthermore, 91% of patients successfully completed SBRT after EUS-guided fiducial delivery. Although fiducials can spontaneously migrate from the initial injection site, the rate of migration is relatively low (7%), and no migration-related complications occurred over the course of this study. Limitations to EUS-guided fiducial placement may include surgically altered anatomy (pancreaticoduodenectomy) in patients with recurrent pancreatic cancer.


Asunto(s)
Endosonografía/métodos , Agujas , Recurrencia Local de Neoplasia/cirugía , Neoplasias Pancreáticas/cirugía , Radiocirugia/instrumentación , Anciano , Progresión de la Enfermedad , Diseño de Equipo , Femenino , Estudios de Seguimiento , Humanos , Imagen por Resonancia Magnética , Masculino , Recurrencia Local de Neoplasia/diagnóstico por imagen , Neoplasias Pancreáticas/diagnóstico por imagen , Estudios Prospectivos , Reproducibilidad de los Resultados , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
19.
Gastrointest Endosc ; 69(6): 1074-80, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19152901

RESUMEN

BACKGROUND: The clinical course of pancreatic endocrine tumors (PET) depends on tumor size, the presence of invasion or metastasis, the Ki-67 index, mitoses per high power field, and mutational damage. Most of this information is not available before surgery for clinical decision making or prognostication. OBJECTIVE: To evaluate PET EUS-guided FNA (EUS-FNA) microsatellite loss analysis in the context of PET-related mortality. DESIGN: A single institution retrospective cohort. PATIENTS: Patients with PET diagnosed by EUS-FNA who underwent DNA microsatellite loss analysis and at least 1 year of follow-up or subsequent death. INTERVENTION: PET microsatellite loss analysis results and current clinical status were compared. RESULTS: Twenty-nine patients were included in the final analysis; the mean age of the patients was 57 years, and 10 were women (35%). The mean follow-up was 33.7 months (median 30 months, range 2-66 months). Twelve patients had disease progression, and 8 died, all from disease-specific causes. Malignant PET contained multiple microsatellite losses, with a median fractional allelic loss (FAL) of 0.37 (range 0.12-0.69, interquartile range [IQR] 0.23-0.42), significantly different from benign PET, median FAL 0 (range 0-0.18, IQR 0-0.08, P < .0001). Survival analysis revealed a significant difference in disease recurrence or progression at 2 years (P < .0001) and in the 5-year survival between patients with FAL 0.2 (P < .0001). Logistic regression could not be performed because of the perfect association between an FAL >0.2 and disease status or mortality. LIMITATIONS: Retrospective design, referral bias, and DNA analysis availability. CONCLUSIONS: PET EUS-FNA microsatellite loss analysis provides preoperative prognostic information. An FAL >0.2 is not only associated with disease progression but also with mortality.


Asunto(s)
Biopsia con Aguja Fina , Carcinoma de Células de los Islotes Pancreáticos/diagnóstico por imagen , Carcinoma de Células de los Islotes Pancreáticos/genética , Endosonografía , Pérdida de Heterocigocidad/genética , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/genética , Ultrasonografía Intervencional , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células de los Islotes Pancreáticos/mortalidad , Carcinoma de Células de los Islotes Pancreáticos/patología , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Estimación de Kaplan-Meier , Antígeno Ki-67/genética , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Páncreas/diagnóstico por imagen , Páncreas/patología , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Pronóstico
20.
Gastroenterol Res Pract ; 2018: 2380596, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29967636

RESUMEN

BACKGROUND: Adolescent obesity is a national epidemic that recently has been shown to increase risk for pancreatic adenocarcinoma (PC) and is associated with an earlier age of PC onset. We hypothesized that PC patients who are overweight or obese at age 18 would have an earlier age of PC onset. METHODS: Retrospective review of 531 patients in our PC registry was completed. Self-reported weight at age 18 and maximum lifetime weight were used to calculate body mass index (BMI) at age 18 (BMI-18) and maximum lifetime BMI. RESULTS: Complete BMI and baseline covariate data was available in 319 PC patients. Mean age (in years) of PC diagnosis for patients whose BMI-18 was overweight (64.0) or obese (59.9) was significantly different when compared to patients with a normal BMI-18 (66.7). No significant difference was observed in the mean age of PC diagnosis in those patients who maintained a normal BMI-18 when compared to those patients who subsequently became overweight or obese (67.0 versus 66.6; p = 0.65). CONCLUSIONS: An elevated BMI at age 18 is associated with an earlier age of PC onset and should be factored into determining the optimal age of beginning screening for patients at high risk for PC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA