Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.759
Filtrar
1.
Front Endocrinol (Lausanne) ; 15: 1281622, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38524630

RESUMO

Background: CXC chemokine receptor 4 (CXCR4) is associated with the progression and metastasis of numerous malignant tumors. However, its relationship with Gastroenteropancreatic Neuroendocrine Neoplasms Grade 3 (GEP-NENs G3) is unclear. The aim of this study was to characterize the expression of CXCR4 in GEP-NENS and to explore the clinical and prognostic value of CXCR4. Methods: This study retrospectively collected clinical and pathological data from patients with GEP-NENs who receiving surgery in Qilu Hospital of Shandong University from January 2013 to April 2021, and obtained the overall survival of the patients based on follow-up. Immunohistochemistry (IHC) was performed on pathological paraffin sections to observe CXCR4 staining. Groups were made according to pathological findings. Kaplan-Meier (K-M) curve was used to evaluate prognosis. SPSS 26.0 was used for statistical analysis. Results: 100 GEP-NENs G3 patients were enrolled in this study. There was a significant difference in primary sites (P=0.002), Ki-67 index (P<0.001), and Carcinoembryonic Antigen (CEA) elevation (P=0.008) between neuroendocrine tumor (NET) G3 and neuroendocrine carcinoma (NEC). CXCR4 was highly expressed only in tumors, low or no expressed in adjacent tissues (P<0.001). The expression level of CXCR4 in NEC was significantly higher than that in NET G3 (P=0.038). The K-M curves showed that there was no significant difference in overall survival between patients with high CXCR4 expression and patients with low CXCR4 expression, either in GEP-NEN G3 or NEC (P=0.920, P=0.842. respectively). Conclusion: Differential expression of CXCR4 was found between tumor and adjacent tissues and between NET G3 and NEC. Our results demonstrated that CXCR4 can be served as a new IHC diagnostic indicator in the diagnosis and differential diagnosis of GEP-NENs G3. Further studies with multi-center, large sample size and longer follow-up are needed to confirm the correlation between CXCR4 expression level and prognosis.


Assuntos
Carcinoma Neuroendócrino , Neoplasias Intestinais , Tumores Neuroendócrinos , Neoplasias Pancreáticas , Neoplasias Gástricas , Humanos , Receptores CXCR4 , Estudos Retrospectivos , Neoplasias Intestinais/patologia , Neoplasias Gástricas/patologia , Neoplasias Pancreáticas/patologia , Tumores Neuroendócrinos/patologia , Carcinoma Neuroendócrino/patologia
5.
Can Vet J ; 65(1): 17-24, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38164388

RESUMO

Two shih tzu dogs were referred to our clinic because of hematochezia and vomiting. Abdominal ultrasonography revealed a focal, asymmetric, exophytic small intestinal mass with loss of wall layering and muscular layer thickening of the adjacent intestine. Computed tomography (CT) in both dogs revealed a focal, asymmetric, homogenously contrast-enhanced exophytic jejunal and duodenal mass with an intact mucosal layer and generalized lymphadenopathy. Metastasis and ulceration were not detected on CT. The initial imaging diagnosis was lymphoma in both dogs; however, histopathological examination revealed the presence of intestinal mast cell tumors (iMCTs). Despite its similarity to alimentary lymphoma, iMCT should be considered a possible diagnosis, based on imaging characteristics, to ensure that proper treatments are selected. This is the first veterinary report describing detailed ultrasonographic and CT characteristics of iMCTs. Key clinical message: This is the first veterinary case report demonstrating sonographic and computed tomographic features of canine iMCT, which can be misdiagnosed as alimentary lymphoma. This report provides another differential diagnosis to consider when determining the appropriate patient treatment direction and histopathological examination.


Caractéristiques échographiques et tomodensitométriques de mastocytomes intestinaux imitant un lymphome alimentaire chez 2 chiens. Deux chiens shih tzu ont été référés à notre clinique en raison d'une hématochézie et de vomissements. L'échographie abdominale a révélé une masse focale, asymétrique et exophytique de l'intestin grêle avec perte de stratification pariétale et épaississement de la couche musculaire de l'intestin adjacent. La tomodensitométrie (TDM) chez les deux chiens a révélé une masse jéjunale et duodénale exophytique focale, asymétrique, homogène et contrastée avec une couche muqueuse intacte et une lymphadénopathie généralisée. Les métastases et les ulcérations n'ont pas été détectées par TDM. Le diagnostic initial d'imagerie était un lymphome chez les deux chiens; cependant, l'examen histopathologique a révélé la présence de mastocytomes intestinaux (iMCT). Malgré sa similitude avec le lymphome alimentaire, l'iMCT doit être considérée comme un diagnostic possible, basé sur les caractéristiques de l'imagerie, afin de garantir la sélection des traitements appropriés. Il s'agit du premier rapport vétérinaire décrivant les caractéristiques échographiques et tomodensitométriques détaillées des iMCT.Message clinique clé :Il s'agit du premier rapport de cas vétérinaire démontrant les caractéristiques échographiques et tomodensitométriques de l'iMCT canin, qui peuvent être diagnostiquées à tort comme un lymphome alimentaire. Ce rapport fournit un autre diagnostic différentiel à prendre en compte lors de la détermination de l'orientation thérapeutique et de l'examen histopathologique appropriés du patient.(Traduit par Dr Serge Messier).


Assuntos
Doenças do Cão , Neoplasias Intestinais , Linfoma , Cães , Animais , Mastócitos/patologia , Neoplasias Intestinais/patologia , Neoplasias Intestinais/veterinária , Linfoma/diagnóstico por imagem , Linfoma/veterinária , Tomografia Computadorizada por Raios X/veterinária , Ultrassonografia/veterinária , Doenças do Cão/diagnóstico por imagem , Doenças do Cão/patologia , Estudos Retrospectivos
6.
J Neuroendocrinol ; 36(2): e13364, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38246597

RESUMO

Neuroendocrine tumors of the small intestine (SI-NETs) often develop lymph node metastasis (LNM)-induced mesenteric fibrosis (MF). MF can cause intestinal obstruction as well as ischemia and render surgical resection technically challenging. The underlying pathomechanisms of MF are still not well understood. We examined mesenteric LNM and the surrounding stroma compartment from 24 SI-NET patients, including 11 with in situ presentation of strong MF (MF+) and 13 without MF (MF-). Differential gene expression was assessed with the HTG EdgeSeq Oncology Biomarker Panel comparing MF+ with MF- within LNM and paired stromal samples, respectively. Most interesting differentially expressed genes were validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) in combination with validation of associated protein levels utilizing immunohistochemistry (IHC) staining of MF+ and MF- formalin-fixed, paraffin-embedded (FFPE) patient samples. Overall, 14 genes measured with a 2549-gene expression panel were differentially expressed in MF+ patients compared to MF-. Of those, nine were differentially expressed genes in LNM and five genes in the stromal tissue (>2-fold change, p < .05). The top hits included increased COMP and COL11A1 expression in the stroma of MF+ patients compared to MF-, as well as decreased HMGA2, COL6A6, and SLC22A3 expression in LNM of MF+ patients compared to LNM of MF- patients. RT-qPCR confirmed high levels of COMP and COL11A1 in stroma samples of MF+ compared to MF- patients. IHC staining confirmed the enrichment of α-smooth muscle actin-positive fibrosis in MF+ compared to MF- patients with corresponding increase of COMP-expressing stromal cells in MF+. Since COMP is associated with the known driver for fibrosis development transforming growth factor beta and with a cancer-associated fibroblasts enriched environment, it seems to be a promising new target for MF research.


Assuntos
Neoplasias Intestinais , Tumores Neuroendócrinos , Humanos , Actinas , Tumores Neuroendócrinos/patologia , Neoplasias Intestinais/patologia , Fibrose , Metástase Linfática/patologia , Células Estromais/patologia , Músculo Liso/patologia
9.
Ann Surg Oncol ; 31(2): 1125-1137, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38006527

RESUMO

Small bowel neuroendocrine tumors (SB-NETs) are increasingly identified and have become the most frequent entity among small bowel tumors. An increasing incidence, a high prevalence, and a prolonged survival with optimal modern multidisciplinary management makes SB-NETs a unique set of tumors to consider for surgical oncologists. The major goals of surgical treatment in the setting of SB-NET include control of tumor volume, control of endocrine secretion, and prevention of locoregional complications. Key considerations include assessment of multifocality and resection of mesenteric nodal masses with the use of mesenteric-sparing approaches and acceptance of R1 margins if necessary to clear disease while avoiding short bowel syndrome. A description through eight steps for consideration is presented to allow for systematic surgical planning and execution of resection. Moreover, some controversies and evolving considerations to the surgical principles and technical procedures remain. The role of primary tumor resection in the presence of (unresectable) liver metastasis is still unclear. Reports of feasibility of minimally invasive surgery are emerging, with undetermined selection criteria for appropriateness or long-term outcomes. Resection of SB-NETs should be considered in all patients fit for surgery and should follow principles to achieve surgical oncological control that is appropriate for the stage and tumor burden, considering the age and comorbidity of the individual patient.


Assuntos
Neoplasias Intestinais , Tumores Neuroendócrinos , Neoplasias Pancreáticas , Humanos , Tumores Neuroendócrinos/patologia , Neoplasias Intestinais/cirurgia , Neoplasias Intestinais/patologia , Neoplasias Pancreáticas/cirurgia , Excisão de Linfonodo
10.
Int J Mol Sci ; 24(24)2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38139230

RESUMO

Determining neuroendocrine tumor (NET) primary sites is pivotal for patient care as pancreatic NETs (pNETs) and small bowel NETs (sbNETs) have distinct treatment approaches. The diagnostic power and prioritization of fluorescence in situ hybridization (FISH) assay biomarkers for establishing primary sites has not been thoroughly investigated using machine learning (ML) techniques. We trained ML models on FISH assay metrics from 85 sbNET and 59 pNET samples for primary site prediction. Exploring multiple methods for imputing missing data, the impute-by-median dataset coupled with a support vector machine model achieved the highest classification accuracy of 93.1% on a held-out test set, with the top importance variables originating from the ERBB2 FISH probe. Due to the greater interpretability of decision tree (DT) models, we fit DT models to ten dataset splits, achieving optimal performance with k-nearest neighbor (KNN) imputed data and a transformation to single categorical biomarker probe variables, with a mean accuracy of 81.4%, on held-out test sets. ERBB2 and MET variables ranked as top-performing features in 9 of 10 DT models and the full dataset model. These findings offer probabilistic guidance for FISH testing, emphasizing the prioritization of the ERBB2, SMAD4, and CDKN2A FISH probes in diagnosing NET primary sites.


Assuntos
Neoplasias Intestinais , Tumores Neuroendócrinos , Neoplasias Pancreáticas , Humanos , Tumores Neuroendócrinos/diagnóstico , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/patologia , Hibridização in Situ Fluorescente , Neoplasias Intestinais/patologia , Neoplasias Pancreáticas/patologia , Aprendizado de Máquina
11.
F1000Res ; 12: 417, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37954063

RESUMO

Background: Small bowel carcinoids are insidious tumors that are often metastatic when diagnosed. Limited mutation landscape studies of carcinoids indicate that these tumors have a relatively low mutational burden. The development of targeted therapies will depend upon the identification of mutations that drive the pathogenesis and metastasis of carcinoid tumors. Methods: Whole exome and RNA sequencing of 5 matched sets of normal tissue, primary small intestine carcinoid tumors, and liver metastases were investigated. Germline and somatic variants included: single nucleotide variants (SNVs), insertions/deletions (indels), structural variants, and copy number alterations (CNAs). The functional impact of mutations was predicted using Ensembl Variant Effect Predictor. Results: Large-scale CNAs were observed including the loss of chromosome 18 in all 5 metastases and 3/5 primary tumors. Certain somatic SNVs were metastasis-specific; including mutations in ATRX, CDKN1B, MXRA5 (leading to the activation of a cryptic splice site and loss of mRNA), SMARCA2, and the loss of UBE4B. Additional mutations in ATRX, and splice site loss of PYGL, leading to intron retention observed in primary and metastatic tumors. Conclusions: We observed novel mutations in primary/metastatic carcinoid tumor pairs, and some have been observed in other types of neuroendocrine tumors. We confirmed a previously observed loss of chromosome 18 and CDKN1B. Transcriptome sequencing added relevant information that would not have been appreciated with DNA sequencing alone. The detection of several splicing mutations on the DNA level and their consequences at the RNA level suggests that RNA splicing aberrations may be an important mechanism underlying carcinoid tumors.


Assuntos
Tumor Carcinoide , Neoplasias Intestinais , Tumores Neuroendócrinos , Humanos , Multiômica , Tumor Carcinoide/genética , Tumor Carcinoide/patologia , Tumor Carcinoide/secundário , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/patologia , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Ubiquitina-Proteína Ligases
12.
Curr Oncol ; 30(10): 9192-9204, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37887564

RESUMO

Small intestinal neuroendocrine tumours (SI-NETs) are the most common small intestinal tumours. A particularly challenging subset of these tumours is those that involve the superior mesenteric artery or vein for which the role and feasibility of surgery are often questioned. This systematic review aimed to identify and evaluate the management strategies used for these complex SI-NETs. The identified studies showed positive outcomes with surgery and multimodality therapy.


Assuntos
Neoplasias Intestinais , Tumores Neuroendócrinos , Humanos , Tumores Neuroendócrinos/patologia , Intestino Delgado/patologia , Intestino Delgado/cirurgia , Neoplasias Intestinais/patologia , Neoplasias Intestinais/cirurgia
13.
Curr Oncol ; 30(10): 9244-9261, 2023 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-37887568

RESUMO

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) account for 80% of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs). GEP-NETs are well-differentiated tumors, highly heterogeneous in biology and origin, and are often diagnosed at the metastatic stage. Diagnosis is commonly through clinical symptoms, histopathology, and PET-CT imaging, while molecular markers for metastasis and the primary site are unknown. Here, we report the identification of multi-gene signatures for hepatic metastasis and primary sites through analyses on RNA-SEQ datasets of pancreatic and small intestinal NETs tissue samples. Relevant gene features, identified from the normalized RNA-SEQ data using the mRMRe algorithm, were used to develop seven Machine Learning models (LDA, RF, CART, k-NN, SVM, XGBOOST, GBM). Two multi-gene random forest (RF) models classified primary and metastatic samples with 100% accuracy in training and test cohorts and >90% accuracy in an independent validation cohort. Similarly, three multi-gene RF models identified the pancreas or small intestine as the primary site with 100% accuracy in training and test cohorts, and >95% accuracy in an independent cohort. Multi-label models for concurrent prediction of hepatic metastasis and primary site returned >98.42% and >87.42% accuracies on training and test cohorts, respectively. A robust molecular signature to predict liver metastasis or the primary site for GEP-NETs is reported for the first time and could complement the clinical management of GEP-NETs.


Assuntos
Neoplasias Intestinais , Neoplasias Hepáticas , Tumores Neuroendócrinos , Humanos , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/patologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Neoplasias Hepáticas/genética
14.
Regul Toxicol Pharmacol ; 145: 105521, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37863416

RESUMO

Hexavalent chromium [Cr(VI)] is present in drinking water from natural and anthropogenic sources at approximately 1 ppb. Several regulatory bodies have recently developed threshold-based safety criteria for Cr(VI) of 30-100 ppb based on evidence that small intestine tumors in mice following exposure to ≥20,000 ppb are the result of a non-mutagenic mode of action (MOA). In contrast, U.S. EPA has recently concluded that Cr(VI) acts through a mutagenic MOA based, in part, on scoring numerous in vivo genotoxicity studies as having low confidence; and therefore derived a cancer slope factor (CSF) of 0.5 (mg/kg-day)-1, equivalent to ∼0.07 ppb. Herein, we demonstrate how physiologically based pharmacokinetic (PBPK) models and intestinal segment-specific tumor incidence data can form a robust dataset supporting derivation of alternative CSF values that equate to Cr(VI) concentrations ranging from below background to concentrations similar to those derived using threshold approaches-depending on benchmark response level and risk tolerance. Additionally, we highlight weaknesses in the rationale EPA used to discount critical in vivo genotoxicity studies. While the data support a non-genotoxic MOA, these alternative toxicity criteria require only PBPK models, robust tumor data, and fair interpretation of published in vivo genotoxicity data for Cr(VI).


Assuntos
Neoplasias Intestinais , Neoplasias Bucais , Camundongos , Animais , Cromo/toxicidade , Neoplasias Intestinais/patologia , Mutagênese , Mutagênicos/toxicidade
15.
PLoS One ; 18(10): e0292643, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37824459

RESUMO

Among the small intestinal tumors that occur in irradiated mice of the established mouse model B6/B6-Chr18MSM-F1 ApcMin/+, loss of heterozygosity analysis can be utilized to estimate whether a deletion in the wild-type allele containing the Adenomatous polyposis coli (Apc) region (hereafter referred to as Deletion), a duplication in the mutant allele with a nonsense mutation at codon 850 of Apc (Duplication), or no aberration (Unidentified) has occurred. Previous research has revealed that the number of Unidentified tumors tends to increase with the radiation dose. In the present study, we investigated the molecular mechanisms underlying the development of an Unidentified tumor type in response to radiation exposure. The mRNA expression levels of Apc were significantly lower in Unidentified tumors than in normal tissues. We focused on epigenetic suppression as the mechanism underlying this decreased expression; however, hypermethylation of the Apc promoter region was not observed. To investigate whether deletions occur that cannot be captured by loss of heterozygosity analysis, we analyzed chromosome 18 using a customized array comparative genomic hybridization approach designed to detect copy-number changes in chromosome 18. However, the copy number of the Apc region was not altered in Unidentified tumors. Finally, gene mutation analysis of the Apc region using next-generation sequencing suggested the existence of a small deletion (approximately 3.5 kbp) in an Unidentified tumor from a mouse in the irradiated group. Furthermore, nonsense and frameshift mutations in Apc were found in approximately 30% of the Unidentified tumors analyzed. These results suggest that radiation-induced Unidentified tumors arise mainly due to decreased Apc expression of an unknown regulatory mechanism that does not depend on promoter hypermethylation, and that some tumors may result from nonsense mutations which are as-yet undefined point mutations.


Assuntos
Polipose Adenomatosa do Colo , Neoplasias Intestinais , Neoplasias Induzidas por Radiação , Camundongos , Animais , Genes APC , Hibridização Genômica Comparativa , Mutação , Polipose Adenomatosa do Colo/genética , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Neoplasias Induzidas por Radiação/genética , Genômica
16.
Appl Immunohistochem Mol Morphol ; 31(10): 668-672, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37751245

RESUMO

OBJECTIVE: To investigate the expression and clinical significance of HepPar1 and GATA-3 in neuroendocrine neoplasms (NENs). MATERIALS AND METHODS: The expression of HepPar1 and GATA-3 in 144 cases of NENs was detected using immunohistochemistry, and the relevant literature was reviewed. RESULTS: HepPar1 was localized in the cytoplasm, and the positive rate of HepPar1 was 6.25% (9/144) in 144 NENs, 9 of which were derived from gastrointestinal and pancreatic NENs, including 4 cases of neuroendocrine tumor, grade 1 (NET G1), 4 cases of NET G2, and 1 case of NET G3. GATA-3 was localized in the nucleus; the positive rate of GATA-3 was 7.62% (11/144), which was derived from 5 cases of gastrointestinal and pancreatic NENs, 2 cases of the lung, 2 cases of the liver, 1 case of the testis, and 1 case of the mediastinum. HepPar1 and GATA-3 were coexpressed in 4 cases: 2 cases of gastric NET G1, 1 case of gastric NET G2, and 1 case of pancreatic NET G3 with liver metastasis. CONCLUSIONS: HepPar1 and GATA-3 can be expressed in NENs, which are potential traps for the pathologic and differential diagnosis of tumors.


Assuntos
Neoplasias Intestinais , Tumores Neuroendócrinos , Neoplasias Pancreáticas , Neoplasias Gástricas , Masculino , Humanos , Tumores Neuroendócrinos/patologia , Neoplasias Intestinais/patologia , Neoplasias Gástricas/metabolismo , Neoplasias Pancreáticas/metabolismo
17.
Cancer Med ; 12(19): 19949-19958, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37737059

RESUMO

INTRODUCTION: Neuroendocrine neoplasms (NEN) are rare and heterogeneous epithelial tumors, occurring throughout the body. For gastroenteropancreatic (GEP)-NEN, rising incidence rates were reported for the last decades, with underlying causes remaining largely unexplained. We evaluated NEN trends stratifying by their histologic subtypes. METHODS: Incident cases of GEP-NEN diagnosed between 2005 and 2019 were retrieved from the prospective, population-based Bavarian Cancer Registry. GEP-NEN were divided in their histologic subtypes, that is, neuroendocrine tumors (NET) G1, NET G2/G3, other NET versus small-cell neuroendocrine carcinoma (NEC), large-cell NEC, and other NEC. We calculated annual age-standardized incidence rates (ASIRs) per 100,000 persons for the total of GEP-NEN, NEN histologic subtypes, and tumor sites. We used an annual percentage change (APC) approach including a joinpoint analysis to investigate NEN incidence trends. RESULTS: ASIR of GEP-NEN rose from 2.2 in 2005 to 4.8 in 2019, characterized by a significant increase until 2012 (APC 2005-2012: 10.1%), followed by modest rise (APC 2012-2019: 1.5%). In the last decade, this increase was mainly driven by the rise of NET G1 and G2/G3, while incidence for NEC declined. Over the study period, ASIR increased significantly for all GEP-sites except the colon. APCs were largest for the stomach, the appendix, the pancreas, and the rectum. CONCLUSIONS: This study found a significant increase in the incidence of GEP-NET. Though this development may partially be attributable to the increased use of advanced detection techniques and changes in NEN classification, further research should also focus on the identification of NEN risk factors.


Assuntos
Carcinoma Neuroendócrino , Neoplasias Intestinais , Tumores Neuroendócrinos , Neoplasias Pancreáticas , Neoplasias Gástricas , Humanos , Estudos Prospectivos , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/patologia , Neoplasias Intestinais/epidemiologia , Neoplasias Intestinais/patologia , Neoplasias Gástricas/epidemiologia , Neoplasias Gástricas/patologia , Carcinoma Neuroendócrino/patologia
19.
Pathol Res Pract ; 248: 154674, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37454491

RESUMO

BACKGROUND: Intestinal neuroendocrine tumours (I-NETs) represent a non-negligible entity among intestinal neoplasms, with metastatic spreading usually present at the time of diagnosis. In this context, effective molecular actionable targets are still lacking. Through transcriptome analysis, we aim at refining the molecular taxonomy of I-NETs, also providing insights towards the identification of new therapeutic vulnerabilities. MATERIALS AND METHODS: A retrospective series of 38 primary sporadic, surgically-resected I-NETs were assessed for transcriptome profiling of 20,815 genes. RESULTS: Transcriptome analysis detected 643 highly expressed genes. Unsupervised hierarchical clustering, differential expression analysis and gene set enriched analysis identified three different tumour clusters (CL): CL-A, CL-B, CL-C. CL-A showed the overexpression of ARGFX, BIRC8, NANOS2, and SSTR4 genes. Its most characterizing signatures were those related to cell-junctions, and activation of mTOR and WNT pathway. CL-A was also enriched in T CD8 + lymphocytes. CL-B showed the overexpression of PCSK1, QPCT, ST18, and TPH1 genes. Its most characterizing signatures were those related to adipogenesis, neuroendocrine metabolism, and splice site machinery-related processes. CL-B was also enriched in T CD4 + lymphocytes. CL-C showed the overexpression of ALB, ANG, ARG1, and HP genes. Its most characterizing signatures were complement/coagulation and xenobiotic metabolism. CL-C was also enriched in M1/2 macrophages. These CL-based differences may have therapeutic implications in refining the management of I-NET patients. At last, we described a specific gene-set for differentiating I-NET from pancreatic NET. DISCUSSION: Our data represent an additional step for refining the molecular taxonomy of I-NET, identifying novel transcriptome subgroups with different biology and therapeutic opportunities.


Assuntos
Neoplasias Intestinais , Tumores Neuroendócrinos , Humanos , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/patologia , Estudos Retrospectivos , Perfilação da Expressão Gênica , Intestinos/patologia , Transcriptoma , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...