Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 420
Filtrar
1.
Cancer Med ; 12(15): 16207-16220, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37387515

RESUMEN

BACKGROUND: Rectal neuroendocrine neoplasms (NENs) are rare neoplasms with limited understanding of its genomic alterations and molecular typing. METHODS: The paraffin-embedded tissue specimens of 38 patients with rectal NENs after surgery were subjected to whole gene sequencing (WGS), and mutation profilings were drawn to identify high-frequency mutation genes, copy-number variations (CNVs), tumor mutation burden (TMB), signal pathways, mutation signatures, DNA damage repair (DDR) genes, and molecular types. The differences of mutated genes and signaling pathways in different pathological grades and metastatic/non-metastatic groups were compared. It helped to search for potential targets. RESULTS: C > T and T > C transitions are the most common base substitutions in rectal NENs. DNA mismatch repair deficiency, DNA base modifications, smoking and exposure to ultraviolet light might play a role in the occurrence of rectal NENs. DAXX, KMT2C, BCL2L1, LTK, MERTK, SPEN, PKN1, FAT3, and LRP2 mutations were found in only low-grade rectal NETs, whereas APC, TP53, NF1, SOX9, and BRCA1 mutations were common in high-grade rectal NECs/MiNENs. These genes helped in distinguishing poorly-differentiated or well-differentiated rectal NENs. Alterations in P53, Wnt and TGFß signaling pathways were more pronounced in rectal NECs and MiNENs. Alterations in Wnt, MAPK and PI3K/AKT signaling pathways promoted metastases. Rectal NENs were classified into two molecular subtypes by cluster analysis based on the mutant genes and signaling pathways combined with clinicopathological features. Patients with mutations in the LRP2, DAXX, and PKN1 gene showed a trend of well-differentiated and early-stage tumors with less metastasis (p = 0.000). CONCLUSIONS: This study evaluated risk factors for regional lymphatic and/or distant metastases, identified high-frequency mutated genes, mutation signatures, altered signaling pathways through NGS. Rectal NENs were divided into two molecular types. This helps to evaluate the likelihood of metastasis, formulate follow-up strategies for patients and provide a target for future research on precision treatment of rectal NENs. PARP inhibitors, MEK inhibitors, mTOR/AKT/PI3K and Wnt signaling pathway inhibitors may be effective drugs for the treatment of metastatic rectal NENs.


Asunto(s)
Tumores Neuroendocrinos , Neoplasias del Recto , Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Neoplasias del Recto/clasificación , Neoplasias del Recto/genética , Neoplasias del Recto/patología , Adhesión en Parafina , Mutación , Tipificación Molecular , Análisis Mutacional de ADN , Estadificación de Neoplasias , Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años
2.
Panminerva Med ; 64(2): 259-264, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35146989

RESUMEN

Neuroendocrine neoplasms (NENs) are a group of disease entities sharing common morphological, ultrastructural and immunophenotypical features, yet with distinct biological behavior and clinical outcome, ranging from benign to frankly malignant. Accordingly, a spectrum of therapeutic options for each single entity is available, including somatostatin analogues (SSA), mTOR-inhibitors, peptide receptor radionuclide therapy (PRRT), non-platinum and platinum chemotherapy. In the last few decades, several attempts have been made to better stratify these lesions refining the pathological classifications, so as to obtain an optimal correspondence between the scientific terminology and, the predictive and prognostic features of each disease subtype, and achieve a global Classification encompassing NENs arising at different anatomical sites. The aim of this review was to analyze, compare and discuss the main features and issues of the latest WHO Classifications of NENs of the lung and the digestive system, in order to point out the strengths and limitations of our current understanding of these complex diseases.


Asunto(s)
Neoplasias del Sistema Digestivo , Neoplasias Pulmonares , Tumores Neuroendocrinos , Neoplasias del Sistema Digestivo/clasificación , Humanos , Pulmón/patología , Neoplasias Pulmonares/clasificación , Tumores Neuroendocrinos/clasificación , Organización Mundial de la Salud
3.
J Endocrinol Invest ; 45(4): 849-857, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35040099

RESUMEN

PURPOSE: Risk factors for sporadic GEP-NENs are still not well defined. To identify the main clinical risk factors represents the aim of this study performed by three Italian referral centers for NENs. METHODS: We performed a retrospective case-control study including 148 consecutive sporadic GEP-NENs and 210 age- and sex-matched controls. We collected data on clinical features, cancer family history and other potential risk factors. RESULTS: Mean age was 58.3 ± 15.8 years; 50% males, primary site was pancreas (50.7%), followed by ileum (22.3%). The 62.8% and 29.1% of cases were G1 and G2, respectively; the 40% had locally advanced or metastatic disease at diagnosis. Independent risk factors for GEP-NENs were: family history of non-neuroendocrine GEP cancer (OR 2.16, 95% CI 1.31-3.55, p = 0.003), type 2 diabetes mellitus (T2DM) (OR 2.5, 95% CI 1.39-4.51, p = 0.002) and obesity (OR 1.88, 95% CI 1.18-2.99, p = 0.007). In the T2DM subjects, metformin use was a protective factor (OR 0.28, 95% CI 0.08-0.93, p = 0.049). T2DM was also associated with a more advanced (OR 2.39, 95% CI 1.05-5.46, p = 0.035) and progressive disease (OR 2.47, 95% CI 1.08-5.34, p = 0.03). Stratifying cases by primary site, independent risk factors for pancreatic NENs were T2DM (OR 2.57, 95% CI 1.28-5.15, p = 0.008) and obesity (OR 1.98, 95% CI 1.11-3.52, p = 0.020), while for intestinal NENs family history of non-neuroendocrine GEP cancer (OR 2.46, 95% CI 1.38-4.38, p = 0.003) and obesity (OR 1.90, 95% CI 1.08-3.33, p = 0.026). CONCLUSION: This study reinforces a role for family history of non-neuroendocrine GEP cancer, T2DM and obesity as independent risk factors for GEP-NENs and suggests a role of metformin as a protective factor in T2DM subjects. If confirmed, these findings could have a significant impact on prevention strategies for GEP-NENs.


Asunto(s)
Neoplasias Intestinales/genética , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Neoplasias Gástricas/genética , Adulto , Anciano , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Femenino , Humanos , Neoplasias Intestinales/clasificación , Neoplasias Intestinales/epidemiología , Italia/epidemiología , Masculino , Anamnesis/estadística & datos numéricos , Persona de Mediana Edad , Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/epidemiología , Neoplasias Pancreáticas/clasificación , Neoplasias Pancreáticas/epidemiología , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Neoplasias Gástricas/clasificación , Neoplasias Gástricas/epidemiología
4.
APMIS ; 130(1): 11-20, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34741788

RESUMEN

The Ki-67 proliferation index (PI) is a prognostic factor in neuroendocrine tumors (NETs) and defines tumor grade. Analysis of Ki-67 PI requires calculation of Ki-67-positive and Ki-67-negative tumor cells, which is highly subjective. To overcome this, we developed a deep learning-based Ki-67 PI algorithm (KAI) that objectively calculates Ki-67 PI. Our study material consisted of NETs divided into training (n = 39), testing (n = 124), and validation (n = 60) series. All slides were digitized and processed in the Aiforia® Create (Aiforia Technologies, Helsinki, Finland) platform. The ICC between the pathologists and the KAI was 0.89. In 46% of the tumors, the Ki-67 PIs calculated by the pathologists and the KAI were the same. In 12% of the tumors, the Ki-67 PI calculated by the KAI was 1% lower and in 42% of the tumors on average 3% higher. The DL-based Ki-67 PI algorithm yields results similar to human observers. While the algorithm cannot replace the pathologist, it can assist in the laborious Ki-67 PI assessment of NETs. In the future, this approach could be useful in, for example, multi-center clinical trials where objective estimation of Ki-67 PI is crucial.


Asunto(s)
Biomarcadores de Tumor , Procesamiento de Imagen Asistido por Computador/métodos , Antígeno Ki-67/metabolismo , Tumores Neuroendocrinos/diagnóstico , Tumores Neuroendocrinos/metabolismo , Patología Clínica/métodos , Algoritmos , Automatización , Proliferación Celular , Aprendizaje Profundo , Pruebas Diagnósticas de Rutina/métodos , Finlandia , Humanos , Tumores Neuroendocrinos/clasificación , Reproducibilidad de los Resultados
5.
J Clin Pathol ; 75(2): 76-84, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34321225

RESUMEN

Breast parenchyma progenitor cells show a high degree of phenotypic plasticity reflected in the wide range of morphology observed in benign and malignant breast tumours. Although there is evidence suggesting that all breast cancer (BC) arises from a common epithelial progenitor or stem cell located at the terminal duct lobular units (TDLUs), BC shows a broad spectrum of morphology with extensive variation in histological type and grade. This is related to the complexity of BC carcinogenesis including initial genetic changes in the cell of origin, subsequent genetic and epigenetic alterations and reprogramming that occur at various stages of BC development and the interplay with the surrounding microenvironment, factors which influence the process of differentiation. Differentiation in BC determines the morphology, which can be measured using histological grade and tumour type. Histological grade, which measures the similarity to the TDLUs, reflects the degree of differentiation whereas tumour type reflects the type of differentiation. Understanding BC phenotypic differentiation facilitates the accurate diagnosis and histological classification of BC with corresponding clinical implications in terms of disease behaviour, prognosis and management plans. In this review, we highlight the potential pathways that BC stem cells follow resulting in the development of different histological types of BC and how knowledge of these pathways impacts our ability to classify BC in diagnostic practice. We also discuss the role of cellular differentiation in producing metaplastic and neuroendocrine carcinomas of the breast and how the latter differ from their counterparts in other organs, with emphasis on clinical relevance.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma/patología , Diferenciación Celular , Células Madre Neoplásicas/patología , Tumores Neuroendocrinos/patología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma/clasificación , Carcinoma/genética , Carcinoma/metabolismo , Linaje de la Célula , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metaplasia , Clasificación del Tumor , Células Madre Neoplásicas/metabolismo , Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/metabolismo , Fenotipo
6.
Nat Commun ; 12(1): 4612, 2021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-34326338

RESUMEN

Metastatic and locally-advanced neuroendocrine neoplasms (aNEN) form clinically and genetically heterogeneous malignancies, characterized by distinct prognoses based upon primary tumor localization, functionality, grade, proliferation index and diverse outcomes to treatment. Here, we report the mutational landscape of 85 whole-genome sequenced aNEN. This landscape reveals distinct genomic subpopulations of aNEN based on primary localization and differentiation grade; we observe relatively high tumor mutational burdens (TMB) in neuroendocrine carcinoma (average 5.45 somatic mutations per megabase) with TP53, KRAS, RB1, CSMD3, APC, CSMD1, LRATD2, TRRAP and MYC as major drivers versus an overall low TMB in neuroendocrine tumors (1.09). Furthermore, we observe distinct drivers which are enriched in somatic aberrations in pancreatic (MEN1, ATRX, DAXX, DMD and CREBBP) and midgut-derived neuroendocrine tumors (CDKN1B). Finally, 49% of aNEN patients reveal potential therapeutic targets based upon actionable (and responsive) somatic aberrations within their genome; potentially directing improvements in aNEN treatment strategies.


Asunto(s)
Mutación , Tumores Neuroendocrinos/genética , Anciano , Femenino , Genes Relacionados con las Neoplasias , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Pronóstico , Secuenciación Completa del Genoma/métodos
7.
Endocrinol Diabetes Nutr (Engl Ed) ; 68(3): 196-207, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34167699

RESUMEN

Pituitary neuroendocrine tumors (PitNETs) constitute, together with other tumors of the sellar region, 15-25% of intracranial neoplasms. In 2017, the World Health Organization proposed a new classification of PitNETs. The main innovation with respect to the 2004 classification was the recommendation to include in the immunohistochemical evaluation of PitNETs the determination of the transcription factors of the 3 pituitary cell lineages: Pit-1, Tpit and SF-1. Additionally, other clinicopathological classifications with a predictive capacity of tumor behavior during follow-up were proposed. Given these changes, it is appropriate to adapt the knowledge generated during the last 15 years to the daily practice of the treatment and monitoring of PitNETs at the Centers of Excellence in Pituitary Pathology. This document includes the positioning of the Spanish Society of Endocrinology and Nutrition (SEEN) and the Spanish Society of Pathology (SEAP) on the classification and denomination of the PitNETs and the information that the pathologist should provide to the clinician to facilitate the treatment and monitoring of these tumors.


Asunto(s)
Tumores Neuroendocrinos , Neoplasias Hipofisarias , Consenso , Humanos , Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/diagnóstico , Hipófisis , Neoplasias Hipofisarias/clasificación , Neoplasias Hipofisarias/diagnóstico , Sociedades Médicas , España
8.
Endocr Pathol ; 32(4): 433-441, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34041698

RESUMEN

This morphological and immunohistochemical study demonstrates that tumors currently known as "middle ear adenomas" are truly well-differentiated epithelial neuroendocrine tumors (NETs) composed of cells comparable to normal intestinal L cells, and therefore, these tumors resemble hindgut NETs. These tumors show consistent expression of glucagon, pancreatic polypeptide, PYY, and the transcription factor SATB2, as well as generic neuroendocrine markers and keratins. The same L cell markers are expressed by cells within the normal middle ear epithelium. These markers define a valuable immunohistochemical profile that can be used for differential diagnosis of middle ear neoplasms, particularly in distinguishing epithelial NETs from paragangliomas. The discovery of neuroendocrine cells expressing the same markers in non-neoplastic middle ear mucosa opens new areas of investigation into the physiology of the normal middle ear and the pathophysiology of middle ear disorders.


Asunto(s)
Adenoma/diagnóstico , Neoplasias del Oído/diagnóstico , Oído Medio/patología , Células L/fisiología , Tumores Neuroendocrinos/diagnóstico , Adenoma/clasificación , Adenoma/metabolismo , Adenoma/patología , Adulto , Anciano , Animales , Diferenciación Celular , Diagnóstico Diferencial , Neoplasias del Oído/clasificación , Neoplasias del Oído/metabolismo , Neoplasias del Oído/patología , Oído Medio/metabolismo , Femenino , Humanos , Inmunohistoquímica , Células L/metabolismo , Células L/patología , Masculino , Ratones , Persona de Mediana Edad , Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Estudios Retrospectivos , Terminología como Asunto
9.
Mod Pathol ; 34(9): 1634-1650, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34017065

RESUMEN

The classification of adenohypophysial neoplasms as "pituitary neuroendocrine tumors" (PitNETs) was proposed in 2017 to reflect their characteristics as epithelial neuroendocrine neoplasms with a spectrum of clinical behaviors ranging from small indolent lesions to large, locally invasive, unresectable tumors. Tumor growth and hormone hypersecretion cause significant morbidity and mortality in a subset of patients. The proposal was endorsed by a WHO working group that sought to provide a unified approach to neuroendocrine neoplasia in all body sites. We review the features that are characteristic of neuroendocrine cells, the epidemiology and prognosis of these tumors, as well as further refinements in terms used for other pituitary tumors to ensure consistency with the WHO framework. The intense study of PitNETs has provided information about the importance of cellular differentiation in tumor prognosis as a model for neuroendocrine tumors in different locations.


Asunto(s)
Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/patología , Neoplasias Hipofisarias/clasificación , Neoplasias Hipofisarias/patología , Humanos
11.
Pancreas ; 50(4): 516-523, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33939663

RESUMEN

OBJECTIVES: There is a pressing need to develop clinical management pathways for grade 3 (G3) gastroenteropancreatic neuroendocrine neoplasms (GEP NEN). METHODS: We performed a retrospective study on patients with metastatic G3 GEP NEN. The relationship between baseline characteristics and progression-free survival and overall survival was analyzed using the Kaplan-Meier method. Univariate and multivariate analyses were performed using the Cox proportional hazards model. RESULTS: We included 142 patients (74 well-differentiated neuroendocrine tumors [WDNETs], 68 poorly differentiated neuroendocrine carcinomas [PDNECs]). Patients with WDNET had prolonged survival compared with PDNEC (median, 24 vs 15 months, P = 0.0001), which persisted in both pancreatic and nonpancreatic cohorts. Well-differentiated morphology, Ki-67 <50% and positive somatostatin receptor imaging were independently associated with prolonged survival. Of the subgroup treated with first-line platinum-based chemotherapy, response rates were favorable (partial response, 47%; stable disease, 30%); there was no significant difference in response rates nor progression-free survival between WDNET and PDNEC despite significantly prolonged overall survival in the WDNET cohort. CONCLUSIONS: Our study corroborates the knowledge of 2 prognostically distinct subgroups within the World Health Organization 2019 G3 GEP NEN population, observed in both pancreatic and nonpancreatic gastrointestinal cohorts. Definitive management pathways are needed to reflect the differences between G3 WDNET and PDNEC.


Asunto(s)
Neoplasias Intestinales/patología , Tumores Neuroendocrinos/patología , Páncreas/patología , Neoplasias Pancreáticas/patología , Neoplasias Gástricas/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Neoplasias Intestinales/clasificación , Neoplasias Intestinales/tratamiento farmacológico , Estimación de Kaplan-Meier , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/tratamiento farmacológico , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Neoplasias Pancreáticas/clasificación , Neoplasias Pancreáticas/tratamiento farmacológico , Pronóstico , Estudios Retrospectivos , Neoplasias Gástricas/clasificación , Neoplasias Gástricas/tratamiento farmacológico , Organización Mundial de la Salud , Adulto Joven
12.
Int J Mol Sci ; 22(8)2021 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-33919851

RESUMEN

Pancreatic neuroendocrine tumors (pNETs) are a rare group of cancers accounting for about 1-2% of all pancreatic neoplasms. About 10% of pNETs arise within endocrine tumor syndromes, such as Multiple Endocrine Neoplasia type 1 (MEN1). pNETs affect 30-80% of MEN1 patients, manifesting prevalently as multiple microadenomas. pNETs in patients with MEN1 are particularly difficult to treat due to differences in their growth potential, their multiplicity, the frequent requirement of extensive surgery, the high rate of post-operative recurrences, and the concomitant development of other tumors. MEN1 syndrome is caused by germinal heterozygote inactivating mutation of the MEN1 gene, encoding the menin tumor suppressor protein. MEN1-related pNETs develop following the complete loss of function of wild-type menin. Menin is a key regulator of endocrine cell plasticity and its loss in these cells is sufficient for tumor initiation. Somatic biallelic loss of wild-type menin in the neuroendocrine pancreas presumably alters the epigenetic control of gene expression, mediated by histone modifications and DNA hypermethylation, as a driver of MEN1-associated pNET tumorigenesis. In this light, epigenetic-based therapies aimed to correct the altered DNA methylation, and/or histone modifications might be a possible therapeutic strategy for MEN1 pNETs, for whom standard treatments fail.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/patología , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Animales , Epigénesis Genética , Humanos , Neoplasia Endocrina Múltiple Tipo 1/clasificación , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/terapia , Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/terapia , Neoplasias Pancreáticas/clasificación , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Transducción de Señal/genética
13.
Curr Oncol Rep ; 23(6): 65, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33855635

RESUMEN

PURPOSE OF REVIEW: Classification and nomenclature of neuroendocrine neoplasms (NEN) have frequently changed over the last years. These changes reflect both increasing knowledge and international standardisation. RECENT FINDINGS: The most recent changes in the Gastro-Entero-Pancreatic system induced the concept of well-differentiated NET with high proliferation rate (NET G3), explaining partially the heterogeneity of G3 NEN. Even if the nomenclature in pulmonary NEN is still different, the terms 'carcinoid' and 'atypical carcinoid' are widely overlapping with NET G1 and NET G2. Molecular data shows an additional heterogeneity both in well-differentiated NET and poorly differentiated NEC. However, no studies are available demonstrating clinical usefulness yet. The heterogeneity of NEN regarding the organ of origin, differentiation and molecular subtypes make development of personalised therapy a challenge needing more international and interdisciplinary collaborations and clinical trials allowing stratification according to biological subgroups.


Asunto(s)
Tumores Neuroendocrinos/patología , Biomarcadores de Tumor , Epigénesis Genética , Humanos , Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/genética , Transcriptoma
14.
Front Endocrinol (Lausanne) ; 12: 604644, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815274

RESUMEN

Pituitary tumors are very complex and heterogeneous and have a very wide range of proliferative and aggressive behaviors, and how to define and classify these tumors remains controversial. This review summarizes the epidemiology and progress in the classification and definition of pituitary tumors, as well as controversial issues. Based on the results of radiologic and autopsy studies, the prevalence of pituitary tumors has recently increased significantly. However, the majority of pituitary tumors are incidentally discovered and asymptomatic, and such tumors are called pituitary incidentalomas. Most of these incidentalomas do not induce symptoms, remain stable in size, and do not need treatment. The recent revised classification strategies mainly depend on immunohistochemistry (IHC) to detect pituitary hormones and pituitary transcription factors; therefore, the accuracy of diagnosing pituitary tumors has improved. Although new classification strategies and definitions for pituitary tumors have been presented, there are still some controversies. The term pituitary neuroendocrine tumor (PitNET) was proposed by the International Pituitary Pathology Club, and this terminology can encompass the unpredictable malignant behavior seen in the subset of aggressive pituitary adenomas (PAs). However, some endocrinologists who oppose this change in terminology have argued that the use of tumor in the terminology is misleading, as it gives PAs a harmful connotation when the majority are not aggressive. Such terminology may add new ambiguity to the origin of PAs and unnecessary anxiety and frustration for the majority of patients with benign PAs. The classification of aggressive PAs mainly relies on subjective judgment of clinical behavior and lacks objective biomarkers and unified diagnostic criteria. However, the term "refractory" could more accurately represent the characteristics of these tumors, including their clinical behaviors, radiological features, and pathologic characteristics. Moreover, the diagnostic criteria for refractory PAs are stricter, more objective, and more accurate than those for aggressive PAs. Early identification of patients with these tumors through recognition and increased awareness of the definition of refractory PAs will encourage the early use of aggressive therapeutic strategies.


Asunto(s)
Neoplasias Hipofisarias/clasificación , Neoplasias Hipofisarias/diagnóstico , Adenoma/clasificación , Adenoma/diagnóstico , Adenoma/epidemiología , Adenoma/patología , Técnicas de Diagnóstico Endocrino/normas , Técnicas de Diagnóstico Endocrino/tendencias , Humanos , Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/diagnóstico , Tumores Neuroendocrinos/epidemiología , Tumores Neuroendocrinos/patología , Neoplasias Hipofisarias/epidemiología , Neoplasias Hipofisarias/patología , Guías de Práctica Clínica como Asunto/normas
15.
Histopathology ; 79(3): 406-415, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33738859

RESUMEN

AIMS: In somatotroph pituitary neuroendocrine tumours (adenomas), a pattern of cytokeratin (CK) 18 expression is used for tumour subclassification, with possible clinical implications. Rare somatotroph tumours do not express CK 18. We aimed to characterise this subset clinically and histologically. METHODS AND RESULTS: Clinical and pathological data for the study were derived from a previously published data set of a cohort of 110 patients with acromegaly. Data included serum levels of insulin-like growth factor 1 (IGF1), growth hormone (GH), prolactin and thyroid-stimulating hormone (TSH), tumour diameter, tumour invasion defined by Knosp grade and immunohistochemical data concerning the expression of Ki67, p53, E-cadherin, somatostatin receptor (SSTR)1, SSTR2A, SSTR3, SSTR5 and D2 dopamine receptor. Additional immunohistochemical analysis (AE1/3, CK 8/18, vimentin, neurofilament light chain, internexin-α) was performed. CK 18 was negative in 10 of 110 (9.1%) tumours. One of these tumours was immunoreactive with CK 8/18 antibody, while the remainder expressed only internexin-α intermediate filament in patterns similar to CK 18 (perinuclear fibrous bodies). CK-negative tumours showed no significant differences with respect to biochemical, radiological or pathological features. They showed significantly higher expression of SSTR2A compared to the sparsely granulated subtype and significantly lower expression of E-cadherin compared to the non-sparsely granulated subtypes of tumours. The tumours showed divergent morphology and hormonal expression: two corresponded to densely granulated tumours and three showed co-expression of prolactin and morphology of either mammosomatotroph or somatotroph-lactotroph tumours. Four tumours showed morphology and immunoprofile compatible with plurihormonal Pit1-positive tumours. CONCLUSIONS: CK-negative somatotroph tumours do not represent a distinct subtype of somatotroph tumours, and can be further subdivided according to their morphology and immunoprofile.


Asunto(s)
Queratina-8/análisis , Tumores Neuroendocrinos , Neoplasias Hipofisarias , Biomarcadores de Tumor/análisis , Tumores Neuroendocrinos/clasificación , Tumores Neuroendocrinos/patología , Hipófisis/patología , Neoplasias Hipofisarias/clasificación , Neoplasias Hipofisarias/patología
16.
Mod Pathol ; 34(6): 1062-1073, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33531618

RESUMEN

Breast cancers with neuroendocrine (NE) differentiation are very heterogeneous, comprising broadly cancers that are morphologically similar to NE tumors (NET) of other anatomic sites, infiltrating breast carcinomas, no special type (IBC-NST) and other special subtypes with NE morphology and/or NE markers expression. Depending on the classification schemes, they are variably included into "NE breast cancers". The latest WHO classification harmonized NE breast cancers with NE neoplasms (NEN) of other organ systems, defined NEN into well-differentiated NET (low Nottingham grade) and poorly-differentiated NE carcinoma (NEC) (high Nottingham grade). Other IBC with NE differentiation are diagnosed based on solely the non-NEN component. Due to the changes in diagnostic criteria, variable results were obtained in the previous studies on NE breast cancers. Hence, the clinical value of NE differentiation in breast cancers is not well investigated and understood. In this review, the current understanding in the pathogenesis, clinical, prognostic, immunhistochemical, and molecular features of "NE breast cancers" is summarized. Controversial issues in their diagnosis and classification are also discussed.


Asunto(s)
Neoplasias de la Mama/patología , Tumores Neuroendocrinos/patología , Neoplasias de la Mama/clasificación , Femenino , Humanos , Tumores Neuroendocrinos/clasificación , Organización Mundial de la Salud
17.
Virchows Arch ; 478(1): 129-135, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33555458

RESUMEN

Neuroendocrine tumors of the thymus (TNET) are exceedingly rare neoplasms. Their histomorphology is identical to neuroendocrine tumors elsewhere in the body (in particular the lungs) and bears no similarity with thymomas and thymic carcinomas. Recent molecular findings have profoundly changed our perception of these tumors and may impact future histological classification systems.


Asunto(s)
Carcinoma Neuroendocrino/clasificación , Tumores Neuroendocrinos/clasificación , Neoplasias del Timo/clasificación , Carcinoma Neuroendocrino/patología , Humanos , Tumores Neuroendocrinos/patología , Timoma/clasificación , Timoma/patología , Neoplasias del Timo/patología
19.
Neuroendocrinology ; 111(4): 320-329, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32097950

RESUMEN

BACKGROUND: In 2014, the World Health Organization (WHO) released a classification system introducing neuroendocrine neoplasms (NENs) of the female reproductive tract, excluding the ovaries. This study aimed to evaluate whether retrospective adaption of the gastroenteropancreatic (GEP)-NEN classification is feasible for ovarian NENs (O-NENs) and correlates with prognosis. METHODS: Sixty-eight patients diagnosed with carcinoid, small cell carcinoma (pulmonary type), paraganglioma, non-small/large cell neuroendocrine carcinoma (NEC), mixed NEC, or undifferentiated carcinomas at 20 institutions in Japan were included in this retrospective cross-sectional study. We identified O-NENs through central pathological review using a common slide set, followed by reclassification according to WHO 2010 guidelines for GEP-NENs. A proportional hazards model was used to assess the association of prognostic factors (age, stage, performance status, histology, and residual disease) with overall survival (OS) and progression-free survival (PFS). RESULTS: Of the 68 enrolled patients, 48 were eligible for analysis. All carcinoids (n = 32) were reclassified as NET G1/G2, whereas 14 of 16 carcinomas were reclassified as NEC/mixed adeno-NEC (MANEC) (Fisher's exact test; p < 0.01). The OS/PFS was 49.0/42.5 months and 6.5/3.9 months for NET G1/G2 and NEC/MANEC, respectively. Histology revealed that NEC/MANEC was associated with increased risk of death (HR = 48.0; 95% CI, 3.93-586; p < 0.01) and disease progression (HR = 51.6; 95% CI, 5.54-480; p < 0.01). CONCLUSION: Retrospective adaption of GEP-NEN classification to O-NENs is feasible and correlates well with the prognosis of O-NENs. This classification could be introduced for ovarian tumors.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias Gastrointestinales/clasificación , Tumores Neuroendocrinos/clasificación , Neoplasias Ováricas/sangre , Neoplasias Ováricas/clasificación , Neoplasias Ováricas/diagnóstico , Neoplasias Pancreáticas/clasificación , Guías de Práctica Clínica como Asunto , Anciano , Estudios Transversales , Femenino , Humanos , Japón , Persona de Mediana Edad , Neoplasias Ováricas/mortalidad , Pronóstico , Estudios Retrospectivos , Organización Mundial de la Salud
20.
Hum Pathol ; 110: 20-30, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33321163

RESUMEN

The immunohistochemistry (IHC) characterization of pituitary transcription factors (PTFs) PIT1, TPIT, and SF1, which enable the identification of three different adenohypophyseal cell lines, has been incorporated into the latest classification system of the World Health Organization (WHO) for pituitary adenomas. This change overturns the concept of the adenoma as solely a hormone producer and classifies these tumors based on their cell lineage. The aim of the study was to provide a diagnostic algorithm, based on IHC expression of hypophyseal hormones with potential use in diagnostic practice, contributing to an improved classification of pituitary adenomas. Our sample included 146 pituitary adenomas previously classified based on hormonal subtypes by IHC (former 2004 WHO criteria) and re-evaluated after the IHC quantification of PIT1, TPIT, and SF1 expression, under WHO 2017 recommendations. We assessed the correlation between expression of PTFs and the classification as per hormonal IHC and correlated clinicopathological profiles based on PTFs. The IHC study of PTFs allowed reclassification of 82% of tumors that were negative for all pituitary hormones, with 21 positive cases for SF1 (reclassified as gonadotroph tumors), 1 positive case for TPIT (reclassified as a corticotroph tumor), and 4 positive cases for PIT1. Using SF1 enabled detection of a substantial portion of gonadotroph tumors, reducing the estimated prevalence of null cell tumors to less than 5%, and identification of plurihormonal pituitary neuroendocrine tumors with PIT1-SF1 coexpression and hormone-negative PIT1s, a group in which we did not observe differences in the clinical behavior compared with the rest of the tumors of the same cell lineage.Our results suggest that applying a diagnostic algorithm based on the study of PTFs could contribute to improving the classification of pituitary adenomas. By adding TPIT assessment, we propose a two-step algorithm, with hypophyseal hormones being used in a selective modality, depending on initial results.


Asunto(s)
Tumores Neuroendocrinos/patología , Neoplasias Hipofisarias/clasificación , Neoplasias Hipofisarias/patología , Factores de Transcripción/metabolismo , Adenoma/metabolismo , Adenoma/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Linaje de la Célula/fisiología , Femenino , Humanos , Inmunohistoquímica/métodos , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/clasificación , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...