Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Eur Radiol ; 33(5): 3416-3424, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36964768

RESUMEN

OBJECTIVES: The recently proposed standardized reporting and data system for somatostatin receptor (SSTR)-targeted PET/CT SSTR-RADS 1.0 showed promising first results in the assessment of diagnosis and treatment planning with peptide receptor radionuclide therapy (PRRT) in neuroendocrine tumors (NET). This study aimed to determine the intra- and interreader agreement of SSTR-RADS 1.0. METHODS: SSTR-PET/CT scans of 100 patients were independently evaluated by 4 readers with different levels of expertise according to the SSTR-RADS 1.0 criteria at 2 time points within 6 weeks. For each scan, a maximum of five target lesions were freely chosen by each reader (not more than three lesions per organ) and stratified according to the SSTR-RADS 1.0 criteria. Overall scan score and binary decision on PRRT were assessed. Intra- and interreader agreement was determined using the intraclass correlation coefficient (ICC). RESULTS: Interreader agreement using SSTR-RADS 1.0 for identical target lesions (ICC ≥ 0.91) and overall scan score (ICC ≥ 0.93) was excellent. The decision to state "functional imaging fulfills requirements for PRRT and qualifies patient as potential candidate for PRRT" also demonstrated excellent agreement among all readers (ICC ≥ 0.86). Intrareader agreement was excellent even among different experience levels when comparing target lesion-based scores (ICC ≥ 0.98), overall scan score (ICC ≥ 0.93), and decision for PRRT (ICC ≥ 0.88). CONCLUSION: SSTR-RADS 1.0 represents a highly reproducible and accurate system for stratifying SSTR-targeted PET/CT scans with high intra- and interreader agreement. The system is a promising approach to standardize the diagnosis and treatment planning in NET patients. KEY POINTS: • SSTR-RADS 1.0 offers high reproducibility and accuracy. • SSTR-RADS 1.0 is a promising method to standardize diagnosis and treatment planning for patients with NET.


Asunto(s)
Tumores Neuroendocrinos , Tomografía Computarizada por Tomografía de Emisión de Positrones , Humanos , Receptores de Somatostatina , Tumores Neuroendocrinos/diagnóstico por imagen , Tumores Neuroendocrinos/terapia , Tumores Neuroendocrinos/patología , Reproducibilidad de los Resultados , Cintigrafía
2.
Pancreatology ; 22(5): 644-650, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35589512

RESUMEN

BACKGROUND: Current guidelines provide weak recommendations to treat small (<2 cm) non-functional pancreatic neuroendocrine tumors with low Ki-67 proliferation index either by resection or clinical follow-up. However, there is a lack of consensus regarding the minimal size of pNET, which allows EUS-guided biopsy with high enough diagnostic accuracy for stratification. METHODS: We conducted a retrospective, bicentric analysis of patients who had undergone EUS-guided pNET sampling in two tertiary care Endoscopy Units in Germany and Poland. Using a recursive partitioning of the tree-aided model, we aimed to stratify the probability of successful EUS-guided biopsy of pNET lesions according to their size and location. RESULTS: In our pNET cohort, successful histological confirmation of a pNET diagnosis was achieved in 59/69 (85.5%) cases at the initial EUS-guided biopsy. In 41 patients with a pNET size less than 18.5 mm, the EUS-guided first biopsy was successful in 90.2%. In 16 of these patients with smaller lesions, EUS-guided sampling was 100% in very small (less than 11 mm) and extremely small lesions (less than 8 mm). The biopsy success rate was 100% in tail lesions in the size range between ≥5.95 and <8.1 mm but only 33.3% independent of the investigator in pancreatic head or body, with an error rate of 11.2% CONCLUSION: Using a recursive partitioning of the tree-aided stratification model, we demonstrate for the first time that in balancing risks and benefits, very small pNETs (<1 cm) in the tail of the pancreas should be sampled under EUS-guidance.


Asunto(s)
Tumores Neuroectodérmicos Primitivos , Tumores Neuroendocrinos , Neoplasias Pancreáticas , Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico , Humanos , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Estudios Retrospectivos
3.
Gut ; 70(9): 1768-1781, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33692095

RESUMEN

Gastroenteropancreatic neuroendocrine neoplasia (GEPNEN) comprises clinically as well as prognostically diverse tumour entities often diagnosed at late stage. Current classification provides a uniform terminology and a Ki67-based grading system, thereby facilitating management. Advances in the study of genomic and epigenetic landscapes have amplified knowledge of tumour biology and enhanced identification of prognostic and potentially predictive treatment subgroups. Translation of this genomic and mechanistic biology into advanced GEPNEN management is limited. 'Targeted' treatments such as somatostatin analogues, peptide receptor radiotherapy, tyrosine kinase inhibitors and mammalian target of rapamycin inhibitors are treatment options but predictive tools are lacking. The inability to identify clonal heterogeneity and define critical oncoregulatory pathways prior to therapy, restrict therapeutic efficacy as does the inability to monitor disease status in real time. Chemotherapy in the poor prognosis NEN G3 group, though associated with acceptable response rates, only leads to short-term tumour control and their molecular biology requires delineation to provide new and more specific treatment options.The future requires an exploration of the NEN tumour genome, its microenvironment and an identification of critical oncologic checkpoints for precise drug targeting. In the advance to personalised medical treatment of patients with GEPNEN, clinical trials need to be based on mechanistic and multidimensional characterisation of each tumour in order to identify the therapeutic agent effective for the individual tumour.This review surveys advances in NEN research and delineates the current status of translation with a view to laying the basis for a genome-based personalised medicine management of advanced GEPNEN.


Asunto(s)
Neoplasias Intestinales/terapia , Tumores Neuroendocrinos/terapia , Neoplasias Pancreáticas/terapia , Medicina de Precisión/métodos , Neoplasias Gástricas/terapia , Biomarcadores de Tumor , Epigénesis Genética , Humanos , Neoplasias Intestinales/diagnóstico , Neoplasias Intestinales/genética , Tumores Neuroendocrinos/diagnóstico , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/genética
4.
Eur J Nucl Med Mol Imaging ; 48(11): 3571-3581, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33928401

RESUMEN

PURPOSE: Radiolabelled somatostatin analogues targeting somatostatin receptors (SSR) are well established for combined positron emission tomography/computer tomography (PET/CT) imaging of neuroendocrine tumours (NET). [18F]SiTATE has recently been introduced showing high image quality, promising clinical performance and improved logistics compared to the clinical reference standard 68Ga-DOTA-TOC. Here we present the first dosimetry and optimal scan time analysis. METHODS: Eight NET patients received a [18F]SiTATE-PET/CT (250 ± 66 MBq) with repeated emission scans (10, 30, 60, 120, 180 min after injection). Biodistribution in normal organs and SSR-positive tumour uptake were assessed. Dosimetry estimates for risk organs were determined using a combined linear-monoexponential model, and by applying 18F S-values and reference target masses for the ICRP89 adult male or female (OLINDA 2.0). Tumour-to-background ratios were compared quantitatively and visually between different scan times. RESULTS: After 1 h, normal organs showed similar tracer uptake with only negligible changes until 3 h post-injection. In contrast, tracer uptake by tumours increased progressively for almost all types of metastases, thus increasing tumour-to-background ratios over time. Dosimetry resulted in a total effective dose of 0.015 ± 0.004 mSv/MBq. Visual evaluation revealed no clinically relevant discrepancies between later scan times, but image quality was rated highest in 60 and 120 min images. CONCLUSION: [18F]SiTATE-PET/CT in NET shows overall high tumour-to-background ratios from 60 to 180 min after injection and an effective dose comparable to 68Ga-labelled alternatives. For clinical use of [18F]SiTATE, the best compromise between image quality and tumour-to-background contrast is reached at 120 min, followed by 60 min after injection.


Asunto(s)
Tumores Neuroendocrinos , Tomografía Computarizada por Tomografía de Emisión de Positrones , Adulto , Computadores , Femenino , Humanos , Masculino , Tumores Neuroendocrinos/diagnóstico por imagen , Tomografía de Emisión de Positrones , Radiometría , Distribución Tisular
5.
BMC Cancer ; 19(1): 893, 2019 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-31492157

RESUMEN

AIMS: To evaluate the short- and long-term outcomes of 3 different endoscopic dissection techniques for upper gastrointestinal (GI) submucosal tumours (SMTs). METHODS: Data for 135 patients withGI SMTs who underwent multiband mucosectomy (MBM), endoscopic submucosal dissection (ESD), or endoscopic submucosal excavation (ESE) were retrospectively assessed. The en bloc resection rate, endoscopic complete resection rate, operation time, potential complications and local recurrence rate were compared. RESULTS: No significant differences were observed in the rate of endoscopic complete resections and pathologic complete resections among the three groups. For SMTs > 15 mm in width, the lowest en bloc resection rate was found for MBM (P = 0.000). MBM was also associated with the shortest procedure time, lowest perforation rate and lowest rate of major bleeding. ESE was the most effective procedure for muscularis propria (MP) lesions but was associated with the longest operation time (P < 0.01). The ESD and ESE groups had similar perforation rates (P > 0.05). No differences were observed in 4-year local recurrence rates among the groups (P = 0.945). CONCLUSIONS: MBM is a simple and effective method for the treatment of small SMTs and achieves clinical success rates similar to those of ESD and ESE. However, ESD and ESE are preferable for larger and deep lesions and are associated with a longer operation time. Nonetheless, all 3 techniques resulted in a low 4-year local recurrence rate. Large-scale randomized clinical trials are needed to further investigate these results.


Asunto(s)
Resección Endoscópica de la Mucosa/métodos , Neoplasias Gastrointestinales/cirugía , Resección Endoscópica de la Mucosa/efectos adversos , Mucosa Esofágica/patología , Mucosa Esofágica/cirugía , Femenino , Estudios de Seguimiento , Mucosa Gástrica/patología , Mucosa Gástrica/cirugía , Neoplasias Gastrointestinales/patología , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Tempo Operativo , Complicaciones Posoperatorias/etiología , Estudios Retrospectivos , Resultado del Tratamiento
6.
BMC Cancer ; 19(1): 575, 2019 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-31196127

RESUMEN

BACKGROUND: Distant metastases frequently occur in gastroenteropancreatic neuroendocrine tumors. If hepatic surgery is not feasible, patients are treated with somatostatin analogs. However, the underlying mechanisms of action of this treatment remain to be defined. The aim of the present study was to analyze the micro-RNA expression profile inter-individually before and after the treatment with somatostatin analogs. MATERIAL AND METHODS: Tumor specimens of all included patients (n = 8) before and after the onset of a therapy with somatostatin analogs were analyzed and a micro-RNA expression profile (754 micro-RNAs) of each probe was generated. This analysis in an intra-individual setting was selected to avoid bias from inter-individual differences. The micro-RNA expression profiles were validated by qPCR. Patients with any other systemic treatment were excluded from the present study. RESULTS: Eight patients were included in the present study of which all had neuroendocrine tumors of the small intestine with diffuse hepatic metastases. Grouped analyses revealed that 15 micro-RNAs were differentially expressed (3 up- and 12 downregulated) after the exposure to somatostatin analogs. Additionally, let-7c-5p and mir-3137 are concordantly regulated in the inter-individually analysis. CONCLUSIONS: This is the first study analyzing the individual micro-RNA expression profile before and after a therapy with somatostatin analogs. Data from this study reveal that somatostatin analogs may in part exert their beneficial effects through an alteration in the micro-RNA expression profile.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Intestinales/tratamiento farmacológico , Intestino Delgado/patología , MicroARNs/genética , Tumores Neuroendocrinos/tratamiento farmacológico , Somatostatina/análogos & derivados , Somatostatina/uso terapéutico , Anciano , Variación Biológica Poblacional , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos
7.
Rev Endocr Metab Disord ; 19(2): 145-158, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29464446

RESUMEN

Neuroendocrine tumors (NETs) are a group of rare and heterogeneous malignancies that can develop in various organs. A significant number of gastroenteropancreatic neuroendocrine tumours (GEP-NETs) is functionally active and presents with symptoms related to the secretion of biologically active substances, leading to the development of distinct clinical syndromes. There are various therapeutic approaches for GEP-NETs, including curative surgery, palliative surgery, local-ablative and loco-regional therapies as well as systemic therapeutic options including peptide receptor radionuclide therapy, cytotoxic therapy, and molecularly targeted therapies. Specific supportive therapy of patients with NETs includes management or prevention of hormone-related clinical syndromes and paraneoplastic states. Supportive therapy plays a key role in NET treatment. Supportive therapy includes debulking surgery and interventional radiologic techniques to reduce tumour bulk or load, as well as systemic medical treatment options to manage or prevent hypersecretion syndromes and treatment-related side effects. Supportive therapies are a type of of comprehensive treatment addressing the patient as a whole person throughout the process of NET treatment. Therefore, supportive therapy also encompasses psychosocial support, expert nursing, nutritional support and management of cancer related pain.


Asunto(s)
Gastrinoma/terapia , Glucagonoma/terapia , Insulinoma/terapia , Neoplasias Intestinales/terapia , Síndrome Carcinoide Maligno/terapia , Tumores Neuroendocrinos/terapia , Cuidados Paliativos/métodos , Neoplasias Pancreáticas/terapia , Síndromes Paraneoplásicos/terapia , Neoplasias Gástricas/terapia , Vipoma/terapia , Humanos
8.
Neuroendocrinology ; 106(1): 58-73, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28226315

RESUMEN

BACKGROUND/AIM: Cyclin-dependent kinases (CDKs) are crucial for cell cycle regulation, and alterations in the cell cycle are often observed in human cancer. CDK4/6 in particular orchestrates G1 phase progression and the G1/S transition. Here, we investigated the in vitro effects of the CDK4/6 inhibitor LEE011 in human neuroendocrine tumor cells. METHODS: The human neuroendocrine tumor cell lines BON1, QGP1, NCI-H727 and GOT1 were treated with different concentrations of LEE011 alone and in combination with 5-fluorouracil and everolimus. RESULTS: Cell viability decreased in a time- and dose-dependent manner in BON1, QGP1, and NCI-H727 cells upon LEE011 treatment, whereas GOT1 cells were treatment resistant. Treatment sensitivity towards LEE011 was associated with the high expression of cyclin D1 and Rb. LEE011 caused the dephosphorylation of Rb and a subsequent G1 phase cell cycle arrest. Combined treatment with LEE011 and 5-fluorouracil or everolimus showed a significant enhancement in the inhibition of cell viability when compared to single-substance treatments due to PI3K-Akt-mTOR and Ras-Raf-MEK-ERK pathway downregulation and cooperative downregulation of cell cycle components. However, LEE011 also exhibited antagonizing effects with 5-fluorouracil, protecting NET cells from DNA-damaging chemotherapy by blocking PARP cleavage and caspase-3/7 activity. CONCLUSIONS: Our data demonstrate that the CDK 4/6 inhibitor LEE011 exhibits promising anti-tumoral properties alone and in combination treatment approaches with 5-fluorouracil or everolimus in human neuroendocrine tumor cell lines.


Asunto(s)
Aminopiridinas/farmacología , Antineoplásicos/farmacología , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Tumores Neuroendocrinos/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/fisiología , Quimioterapia Combinada , Everolimus/farmacología , Fluorouracilo/farmacología , Humanos , Tumores Neuroendocrinos/enzimología , Factores de Tiempo
9.
Neuroendocrinology ; 106(1): 1-19, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-27871087

RESUMEN

BACKGROUND/AIMS: The tumor suppressor p53 is depleted in many tumor cells by the E3 ubiquitin ligase mouse double minute 2 homolog (MDM2) through MDM2/p53 interaction. A novel target for inhibiting p53 degradation and for causing reexpression of p53wild type is inhibition of MDM2. The small molecule NVP-CGM097 is a novel MDM2 inhibitor. We investigated MDM2 inhibition as a target in neuroendocrine tumor cells in vitro. METHODS: Human neuroendocrine tumor cell lines from the pancreas (BON1), lung (NCI-H727), and midgut (GOT1) were incubated with the MDM2 inhibitor NVP-CGM097 (Novartis) at concentrations from 4 to 2,500 nM. RESULTS: While p53wild type GOT1 cells were sensitive to NVP-CGM097, p53mutated BON1 and p53mutated NCI-H727 cells were resistant to NVP-CGM097. Incubation of GOT1 cells with NVP-CGM097 at 100, 500, and 2,500 nM for 96 h caused a significant decline in cell viability to 84.9 ± 9.2% (p < 0.05), 77.4 ± 6.6% (p < 0.01), and 47.7 ± 9.2% (p < 0.01). In a Western blot analysis of GOT1 cells, NVP-CGM097 caused a dose-dependent increase in the expression of p53 and p21 tumor suppressor proteins and a decrease in phospho-Rb and E2F1. Experiments of co-incubation of NVP-CGM097 with 5-fluorouracil, temozolomide, or everolimus each showed additive antiproliferative effects in GOT1 cells. NVP-CGM097 and 5-fluorouracil increased p53 and p21 expression in an additive manner. CONCLUSIONS: MDM2 inhibition seems a promising novel therapeutic target in neuroendocrine tumors harboring p53wild type. Further investigations should examine the potential role of MDM2 inhibitors in neuroendocrine tumor treatment.


Asunto(s)
Antineoplásicos/farmacología , Fluorouracilo/farmacología , Isoquinolinas/farmacología , Tumores Neuroendocrinos/tratamiento farmacológico , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Quimioterapia Combinada , Factor de Transcripción E2F1/metabolismo , Humanos , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína de Retinoblastoma/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
10.
Neuroendocrinology ; 106(4): 335-351, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28968593

RESUMEN

INTRODUCTION: Glycogen synthase kinase 3α/ß (GSK3α/ß) is a serine/threonine kinase that plays a critical role in cancer. AIMS: In this study, we evaluated the effects of the specific GSK3α/ß inhibitor AR-A014418 in vitro to gain novel insights into GSK3α/ß signaling in neuroendocrine tumors (NETs). MATERIALS AND METHODS: Human NET cell lines (BON1, QGP1, H727, and GOT1) were treated with different concentrations of AR-A014418 alone and in combination with lovastatin, everolimus, 5-fluorouracil (5-FU), and γ-irradiation. RESULTS: AR-A014418 significantly dose- and time-dependently decreased cell viability in all 4 NET cell lines through inhibition of epithelial growth factor receptor and mTORC1/p70S6K signaling, as well as cyclin D3 downregulation and induction of pChk1. In all cell lines tested, FACS analysis showed an AR-A014418-induced increase in the sub-G1 phase, reflecting cell death. Apoptosis induction was observed in H727, GOT1 and QGP1 cells, but not in BON1 cells. Furthermore, significant antimigratory effects upon GSK3α/ß inhibition were found and were associated with ß-catenin downregulation in all cell lines tested. Compensatory upregulation of pAkt and pERK in response to GSK3α/ß inhibition was prevented by combining AR-A014418 with the ERK and Akt inhibitor lovastatin. Accordingly, the lovastatin/AR-A014418 combination was synergistic in BON1 and QGP1 cells. Moreover, AR-A014418 displayed promising chemosensitizing effects on 5-FU in QGP1 and slight radiosensitizing properties in BON1 and QGP1 cells. CONCLUSION: Our data provide new insights into the role of GSK3α/ß in NETs and suggest that GSK3α/ß inhibition could be a novel therapeutic option in NETs, especially in combination with lovastatin or 5-FU, depending on tumor entity.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/metabolismo , Tumores Neuroendocrinos/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Tiazoles/farmacología , Urea/análogos & derivados , Urea/farmacología
11.
Eur Radiol ; 27(4): 1676-1684, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27436022

RESUMEN

OBJECTIVES: To quantify the additional value of 68Ga-DOTA-TATE PET/CT in comparison with contrast-enhanced CT alone for primary tumour detection in neuroendocrine cancer of unknown primary (CUP-NET). METHODS: In total, 38 consecutive patients (27 men, 11 women; mean age 62 years) with histologically proven CUP-NET who underwent a contrast-enhanced 68Ga-DOTA-TATE PET/CT scan for primary tumour detection and staging between 2010 and 2014 were included in this IRB-approved retrospective study. Two blinded readers independently analysed the contrast-enhanced CT and 68Ga-DOTA-TATE PET datasets separately and noted from which modality they suspected a primary tumour. Consensus was reached if the results were divergent. Postoperative histopathology (24 patients) and follow-up 68Ga-DOTA-TATE PET/CT imaging (14 patients) served as the reference standards and statistical measures of diagnostic accuracy were calculated accordingly. RESULTS: The majority of confirmed primary tumours were located in the abdomen (ileum in 19 patients, pancreas in 12, lung in 2, small pelvis in 1). High interobserver agreement was noted regarding the suspected primary tumour site (Cohen's k 0.90, p < 0.001). 68Ga-DOTA-TATE PET demonstrated a significantly higher sensitivity (94 % vs. 63 %, p = 0.005) and a significantly higher accuracy (87 % vs. 68 %, p = 0.003) than contrast-enhanced CT. CONCLUSIONS: Ga-DOTA-TATE PET/CT compared with contrast-enhanced CT alone provides an improvement in sensitivity of 50 % and an improvement in accuracy of 30 % in primary tumour detection in CUP-NET. KEY POINTS: • 68Ga-DOTA-TATE PET augments the sensitivity of contrast-enhanced CT by 50 % • 68Ga-DOTA-TATE PET augments the accuracy of contrast-enhanced CT by 30 % • Somatostatin receptor-targeted hybrid imaging optimizes primary tumour detection in CUP-NET.


Asunto(s)
Neoplasias Primarias Desconocidas/diagnóstico por imagen , Tumores Neuroendocrinos/diagnóstico por imagen , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Variaciones Dependientes del Observador , Compuestos Organometálicos , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Tomografía de Emisión de Positrones/métodos , Radiofármacos , Estudios Retrospectivos , Sensibilidad y Especificidad , Tomografía Computarizada por Rayos X/métodos
12.
Neuroendocrinology ; 103(3-4): 383-401, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26338447

RESUMEN

BACKGROUND/AIMS: The hepatocyte growth factor/transmembrane tyrosine kinase receptor c-Met has been defined as a potential target in antitumoral treatment of various carcinomas. We aimed to investigate the direct effect of c-Met inhibition on neuroendocrine tumor cells in vitro. METHODS: The effects of the multi-tyrosine kinase inhibitors cabozantinib and tivantinib and of the highly specific c-Met inhibitor INC280 were investigated in human pancreatic neuroendocrine BON1, bronchopulmonary NCI-H727 and midgut GOT1 cells in vitro. RESULTS: INC280, cabozantinib and tivantinib inhibited c-Met phosphorylation, respectively. However, while equimolar concentrations (10 µM) of cabozantinib and tivantinib inhibited cell viability and cell migration, INC280 had no inhibitory effect. Knockdown experiments with c-Met siRNA also did not demonstrate effects on cell viability. Cabozantinib and tivantinib caused a G2 arrest in neuroendocrine tumor cells. CONCLUSIONS: Our in vitro data suggest that c-Met inhibition alone is not sufficient to exert direct antitumoral or antimigratory effects in neuroendocrine tumor cells. The multi-tyrosine kinase inhibitors cabozantinib and tivantinib show promising antitumoral and antimigratory effects in neuroendocrine tumor cells, which are most probably 'off-target' effects, not mediated by c-Met.


Asunto(s)
Anilidas/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Piridinas/farmacología , Pirrolidinonas/farmacología , Quinolinas/farmacología , Benzamidas , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Receptores ErbB/metabolismo , Citometría de Flujo , Humanos , Imidazoles/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Tumores Neuroendocrinos/patología , Proteína Oncogénica v-akt/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Triazinas/farmacología
13.
Ann Surg Oncol ; 22(1): 164-71, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25190113

RESUMEN

BACKGROUND: Resection is the only curative treatment in patients suffering from neuroendocrine tumors (NETs) of the ileum or the pancreas. Accurate preoperative imaging is critical for surgical planning, as even findings of small and distant metastases may profoundly influence surgical management. METHODS: (68)Ga-DOTATATE PET/CT was performed preoperatively in 44 patients suffering from NET of the ileum (n = 26) or the pancreas (n = 18) before surgery at our University Hospital. Data were analyzed retrospectively by an interdisciplinary team of nuclear medicine and visceral surgery specialists. Intended surgical management was documented before and after availability of PET/CT findings. The team judged whether the imaging findings provided additional information relevant to surgical planning. RESULTS: Imaging results altered surgical management in 9 of 44 (20 %) patients, more specifically in 3 of 26 (12 %) patients with NET of the ileum and in 6 of 18 (33 %) patients with NET of the pancreas. PET/CT findings led to a more invasive surgical approach in 6 cases (3 each of ileum and pancreas) and to a less invasive strategy in 3 patients with NET of the pancreas. Although PET/CT results did not alter management in 35 of 44 patients, somatostatin receptor imaging still provided additional information for surgery planning in more than 95 % of the cases. CONCLUSIONS: Additional information provided by (68)Ga-DOTATATE PET/CT in the preoperative workup significantly influences surgical management in one-fifth of our NET patients and, notably, one-third of those suffering from NET of the pancreas.


Asunto(s)
Radioisótopos de Galio , Neoplasias del Íleon/diagnóstico , Tumores Neuroendocrinos/diagnóstico , Compuestos Organometálicos , Neoplasias Pancreáticas/diagnóstico , Tomografía de Emisión de Positrones/métodos , Tomografía Computarizada por Rayos X/métodos , Adulto , Anciano , Manejo de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Imagen Multimodal , Estadificación de Neoplasias , Pronóstico , Estudios Retrospectivos
14.
J Magn Reson Imaging ; 41(2): 361-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24446275

RESUMEN

PURPOSE: To evaluate the association of therapy-related changes in imaging parameters with progression-free survival (PFS) of patients with unresectable liver metastases from neuroendocrine tumors (NETLMs). MATERIALS AND METHODS: Forty-five radioembolized patients (median age: 62 years; range: 43-75) received a pre- and 3 months posttherapeutic magnetic resonance imaging (MRI) examination. The latter were evaluated for tumor size, arterial enhancement, and necrosis pattern. Influences of therapy-related changes on PFS were analyzed. Statistical analysis included Student's t-test, Wilcoxon test, Cox regression analysis, and Kaplan-Meier curves. RESULTS: The median percentage decrease in sum of diameters was 9.7% (range: 43.9% decrease to 15.4% increase). Twenty-one patients (47%) showed increased necrosis. Three parameters were associated with significantly longer PFS: a decrease of diameter (hazard ratio [HR]: 0.206; 95% confidence interval [CI]: 0.058-0.725; P = 0.0139), a decrease in tumor arterial enhancement (HR: 0.143; 95% CI: 0.029-0.696; P = 0.0160), and an increase in necrosis after 3 months (HR: 0.321; 95% CI: 0.104-0.990; P = 0.0480). Multivariate analysis revealed that changes in diameter and arterial enhancement have complementary information and are associated independently with long PFS. CONCLUSION: A decrease both in sum of diameters and arterial enhancement of metastases, as well as an increase in necrosis, are associated with significantly longer PFS after radioembolization.


Asunto(s)
Embolización Terapéutica/métodos , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Imagen por Resonancia Magnética/métodos , Tumores Neuroendocrinos/patología , Adulto , Anciano , Medios de Contraste , Supervivencia sin Enfermedad , Femenino , Gadolinio DTPA , Humanos , Interpretación de Imagen Asistida por Computador , Masculino , Microesferas , Persona de Mediana Edad , Clasificación del Tumor , Estudios Retrospectivos , Resultado del Tratamiento , Radioisótopos de Itrio/uso terapéutico
15.
Mol Imaging ; 13: 1-10, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24824963

RESUMEN

We investigated the use of Ga-68-DOTA-Tyr3-octreotate (Ga-68-DOTA-TATE) positron emission tomography (PET) and standardized uptake values (SUVs) to predict the effectiveness of treatment with the somatostatin analogue octreotide acetate (Sandostatin LAR) in patients with neuroendocrine tumors (NETs). Thirty patients with well-differentiated NETs of the ileum (grades G1 and G2) were studied with Ga-68-DOTA-TATE. The average SUV of a 50% isocontour volume of interest covering the lesion with maximum uptake (SUV mean) and the maximum SUV (SUV max) were determined. Patients were followed up, and the time to progression was recorded. Twenty-one patients showed progressive disease at the end of the study; nine patients had stable disease. The median progression-free survival (PFS) was 51.0 weeks (95% confidence interval [CI] 26.4-75.6). A cutoff for the SUV max of 29.4 and for the SUV mean of 20.3 could separate between patients with a long PFS (69.0 weeks; 95% CI 9.8-128.2) and a short PFS (26.0 weeks; 95% CI 8.7-43.3) response to octreotide acetate therapy. Patients with high radiotracer uptake had significantly higher PFS with a 2.9-fold higher chance for stable disease after 45 weeks; however, the prognostic performance of SUV max on an individual basis was poor, with a sensitivity of 75% and a specificity of 64%. SUV max and SUV mean of NET tumor lesions in Ga-68-DOTA-TATE PET are important prognostic indices for predicting the response to therapy with octreotide acetate.


Asunto(s)
Neoplasias del Íleon/diagnóstico por imagen , Neoplasias del Íleon/radioterapia , Tumores Neuroendocrinos/diagnóstico por imagen , Tumores Neuroendocrinos/radioterapia , Octreótido/uso terapéutico , Radiofármacos/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Compuestos Heterocíclicos , Humanos , Neoplasias del Íleon/patología , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/patología , Compuestos Organometálicos , Tomografía de Emisión de Positrones , Sensibilidad y Especificidad , Análisis de Supervivencia , Resultado del Tratamiento
16.
Radiology ; 270(2): 517-25, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24056402

RESUMEN

PURPOSE: To evaluate diagnostic performance of gallium 68-tetraazacyclododecane tetraacetic acid-octreotate ((68)Ga-DOTATATE) in detection of recurrent neuroendocrine tumors (NETs). MATERIALS AND METHODS: Approval was waived by the local ethics committee for this retrospective study. Between 2007 and 2011, 63 patients (mean age, 58 years) were examined with (68)Ga-DOTATATE positron emission tomography (PET)/computed tomography (CT) after primary NET curative resection. Reasons for PET/CT were regular follow-up examinations (n = 30), increased plasma levels of tumor markers (n = 27), or clinical suspicion of recurrence (n = 6). Final diagnosis was determined with histopathologic verification (n = 25) or clinical follow-up (n = 38). PET/CT scans were evaluated in consensus by two readers without blinding to clinical information and independently by two readers with blinding. Sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) were calculated. RESULTS: Final diagnosis of NET recurrence was determined in 29 patients. In three other patients, tumors of nonneuroendocrine origin were diagnosed. (68)Ga-DOTATATE PET/CT helped identify NET recurrence in 26 of 29 patients (sensitivity, 90%) and exclude presence of recurrent NET in 28 of 34 patients (specificity, 82% ). PET/CT provided false-positive and false-negative results in six and three patients (PPV, 81% [26 of 32]; NPV, 90% [28 of 31]; accuracy, 86% [54 of 63]). In gastroenteropancreatic NET (n = 45), sensitivity was 94% (17 of 18); specificity was 89% (24 of 27); PPV was 85% (17 of 20); NPV was 96% (24 of 25); and accuracy was 91% (41 of 45). Two blinded readers achieved sensitivity of 79% (23 of 29) and 76% (22 of 29); specificity of 85% (29 of 34) and 94% (32 of 34) (κ = 0.80); and accuracy of 83% and 86%. CONCLUSION: (68)Ga-DOTATATE PET/CT is accurate in detection of recurrent NET. Blinded PET/CT review markedly decreased sensitivity, underlining importance of considering clinical parameters in NET recurrence. Present results must be further validated to substantiate use of (68)Ga-DOTATATE PET/CT in routine follow-up after curative resection of NET.


Asunto(s)
Imagen Multimodal , Recurrencia Local de Neoplasia/diagnóstico por imagen , Tumores Neuroendocrinos/diagnóstico por imagen , Compuestos Organometálicos , Tomografía de Emisión de Positrones , Tomografía Computarizada por Rayos X , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/cirugía , Valor Predictivo de las Pruebas , Estudios Retrospectivos , Sensibilidad y Especificidad
17.
J Magn Reson Imaging ; 40(2): 457-66, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24347148

RESUMEN

PURPOSE: To evaluate the diagnostic accuracy of dynamic-contrast-enhanced (DCE) MRI in comparison to both (18)F-FDG- and (68)Ga-DOTATATE-PET/CT in patients with liver metastases of neuroendocrine neoplasms (NEN). MATERIALS AND METHODS: Thirty-two patients with hepatic metastases from NEN were examined both in DCE-MRI and positron emission tomography/computed tomography (PET/CT), using either (18)F-fluorodeoxyglucose ((18)F-FDG) or (68)Ga-DOTATATE as tracer. DCE-MRI was performed at 3 Tesla with Gd-EOB-DTPA acquiring 48 slices every 2.2 s for 5 min. Three regions of interest (ROIs) representing liver background and liver metastases were defined in fat-saturated T1w three-dimensional GRE MRI sequences in the hepatobiliary phase. Corresponding ROIs were then defined in the DCE-MRI- and in the PET/CT-dataset. Area under the curve (AUC) was calculated for the differentiation between metastases and liver background for DCE-MRI and PET-CT parameters. RESULTS: AUC was very high for SUVmean (mean standardized uptake value) derived from (68)Ga-DOTATATE- (AUC = 0.966), and (18)F-FDG-PET/CT (AUC = 0.989). For DCE-MRI parameters, arterial flow fraction and intracellular uptake fraction showed the highest AUCs (AUC = 0.826, AUC = 0.819, respectively). The combination of those two had an AUC of 0.949. The combination of DCE-MRI and PET-CT parameters resulted in the highest AUC. CONCLUSION: Both PET/CT parameters and DCE-MRI perfusion parameters show a high diagnostic accuracy in the distinction between liver metastases and liver tissue. Our data suggest that both modalities provide complementary information.


Asunto(s)
Gadolinio DTPA , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/secundario , Imagen por Resonancia Magnética/métodos , Tumores Neuroendocrinos/diagnóstico , Tomografía de Emisión de Positrones/métodos , Tomografía Computarizada por Rayos X/métodos , Adulto , Anciano , Anciano de 80 o más Años , Medios de Contraste , Femenino , Fluorodesoxiglucosa F18 , Humanos , Masculino , Persona de Mediana Edad , Imagen Multimodal/métodos , Compuestos Organometálicos , Radiofármacos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
18.
Radiologie (Heidelb) ; 64(7): 559-567, 2024 Jul.
Artículo en Alemán | MEDLINE | ID: mdl-38789854

RESUMEN

BACKGROUND: Neuroendocrine tumors of the pancreas have a broad biological spectrum. The treatment decision is based on an optimal diagnosis with regard to the local findings and possible locoregional and distant metastases. In addition to purely morphologic imaging procedures, functional parameters are playing an increasingly important role in imaging. OBJECTIVES: Prerequisites for optimal imaging of the pancreas, technical principles are provided, and the advantages and disadvantages of common cross-sectional imaging techniques as well as clinical indications for these special imaging methods are discussed. MATERIALS AND METHODS: Guidelines, basic and review papers will be analyzed. RESULTS: Neuroendocrine tumors of the pancreas have a broad imaging spectrum. Therefore, there is a need for multimodality imaging in which morphologic and functional techniques support each other. While positron emission tomography/computed tomography (PET/CT) can determine the presence of one or more lesions and its/their functional status of the tumor, magnetic resonance imaging (MRI) efficiently identifies the location, relationship to the main duct and the presence of liver metastases. CT allows a better vascular evaluation, even in the presence of anatomical variants as well as sensitive detection of lung metastases. CONCLUSIONS: Knowledge of the optimal combination of imaging modalities including clinical and histopathologic results and dedicated imaging techniques is essential to achieve an accurate diagnosis to optimize treatment decision-making and to assess therapy response.


Asunto(s)
Imagen por Resonancia Magnética , Tumores Neuroendocrinos , Neoplasias Pancreáticas , Humanos , Imagen por Resonancia Magnética/métodos , Imagen Multimodal/métodos , Tumores Neuroendocrinos/diagnóstico por imagen , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/secundario , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/patología , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Tomografía Computarizada por Rayos X/métodos
19.
Radiol Oncol ; 58(2): 196-205, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38613843

RESUMEN

BACKGROUND: This study explores the predictive and monitoring capabilities of clinical and multiparametric MR parameters in assessing capecitabine and temozolomide (CAPTEM) therapy response in patients with neuroendocrine tumors (NET). PATIENTS AND METHODS: This retrospective study (n = 44) assessed CAPTEM therapy response in neuroendocrine liver metastases (NELM) patients. Among 33 monitored patients, as a subgroup of the overall study cohort, pretherapeutic and follow-up MRI data (size, apparent diffusion coefficient [ADC] values, and signal intensities), along with clinical parameters (chromogranin A [CgA] and Ki-67%), were analyzed. Progression-free survival (PFS) served as the reference. Responders were defined as those with PFS ≥ 6 months. RESULTS: Most patients were male (75%) and had G2 tumors (76%) with a pancreatic origin (84%). Median PFS was 5.7 months; Overall Survival (OS) was 25 months. Non-responders (NR) had higher Ki-67 in primary tumors (16.5 vs. 10%, p = 0.01) and increased hepatic burden (20% vs. 5%, p = 0.007). NR showed elevated CgA post-treatment, while responders (R) exhibited a mild decrease. ADC changes differed significantly between groups, with NR having decreased ADCmin (-23%) and liver-adjusted ADCmean/ADCmean liver (-16%), compared to R's increases of ADCmin (50%) and ADCmean/ADCmean liver (30%). Receiver operating characteristic (ROC) analysis identified the highest area under the curve (AUC) (0.76) for a single parameter for ∆ ADC mean/liver ADCmean, with a cut-off of < 6.9 (76% sensitivity, 75% specificity). Combining ∆ Size NELM and ∆ ADCmin achieved the best balance (88% sensitivity, 60% specificity) outperforming ∆ Size NELM alone (69% sensitivity, 65% specificity). Kaplan-Meier analysis indicated significantly longer PFS for ∆ ADCmean/ADCmean liver < 6.9 (p = 0.024) and ∆ Size NELM > 0% + ∆ ADCmin < -2.9% (p = 0.021). CONCLUSIONS: Survival analysis emphasizes the need for adapted response criteria, involving combined evaluation of CgA, ADC values, and tumor size for monitoring CAPTEM response in hepatic metastasized NETs.


Asunto(s)
Capecitabina , Neoplasias Hepáticas , Tumores Neuroendocrinos , Temozolomida , Humanos , Temozolomida/uso terapéutico , Temozolomida/administración & dosificación , Masculino , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/diagnóstico por imagen , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/diagnóstico por imagen , Tumores Neuroendocrinos/patología , Femenino , Capecitabina/administración & dosificación , Capecitabina/uso terapéutico , Persona de Mediana Edad , Estudios Retrospectivos , Anciano , Adulto , Imagen por Resonancia Magnética/métodos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resultado del Tratamiento , Antígeno Ki-67/análisis , Antígeno Ki-67/metabolismo , Supervivencia sin Progresión
20.
Radiol Oncol ; 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38861687

RESUMEN

BACKGROUND: This study aimed to assess 68Ga-DOTA-TATE (-TOC) PET/CT quantitative parameters in monitoring and predicting everolimus response in neuroendocrine tumor (NET) patients with hepatic metastases (NELM). PATIENTS AND METHODS: This retrospective analysis included 29 patients with 62 target lesions undergoing everolimus treatment and pre-therapy, and follow-up 68Ga-DOTA-TATE (-TOC) PET/CT scans. Response evaluation utilized progression-free survival (PFS) categorized as responders (R; PFS > 6 months) and non-responders (NR; PFS ≤ 6 months). Lesion size and density, along with maximum and median standardize uptake value (SUV) in target lesions, liver, and spleen were assessed. Tumor-to-spleen (T/S) and tumor-to-liver (T/L) ratios were calculated, including the tumor-to-spleen (T/S) ratio and tumor-to-liver (T/L) ratio (using SUVmax/SUVmax, SUVmax/SUVmean, and SUVmean/SUVmean). RESULTS: PET/CT scans were acquired 19 days (interquartile range [IQR] 69 days) pre-treatment and 127 days (IQR 74 days) post-starting everolimus. The overall median PFS was 264 days (95% CI: 134-394 days). R exhibited significant decreases in Tmax/Lmax and Tmean/Lmax ratios compared to NR (p = 0.01). In univariate Cox regression, Tmean/Lmax ratio was the sole prognostic parameter associated with PFS (HR 0.5, 95% CI 0.28-0.92, p = 0.03). Percentage changes in T/L and T/S ratios were significant predictors of PFS, with the highest area under curve (AUC) for the percentage change of Tmean/Lmax (AUC = 0.73). An optimal threshold of < 2.5% identified patients with longer PFS (p = 0.003). No other imaging or clinical parameters were predictive of PFS. CONCLUSIONS: This study highlights the potential of quantitative SSTR-PET/CT in predicting and monitoring everolimus response in NET patients. Liver metastasis-to-liver parenchyma ratios outperformed size-based criteria, and Tmean/Lmax ratio may serve as a prognostic marker for PFS, warranting larger cohort investigation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA