Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Neuroendocrinology ; 109(2): 83-99, 2019.
Article in English | MEDLINE | ID: mdl-30699437

ABSTRACT

Neuroendocrine tumors (NETs) include a heterogeneous group of malignancies arising in the diffuse neuroendocrine system and characterized by indolent growth. Complex interactions take place among the cellular components of the microenvironment of these tumors, and the recognition of the molecular mediators of their interplay and cross talk is crucial to discover novel therapeutic targets. NET cells overexpress a plethora of proangiogenic molecules including vascular endothelial growth factor, platelet-derived growth factor, fibroblast growth factor, semaphorins, and angiopoietins that promote both recruitment and proliferation of endothelial cell precursors, thus resulting among the most vascularized cancers with a microvessel density 10-fold higher than epithelial tumors. Also, NETs operate multifaceted interactions with stromal cells, both at local and distant sites, and whether their paracrine secretion of serotonin, connective tissue growth factor, and transforming growth factor ß primarily drives the fibroblast activation to enhance the tumor proliferation, on the other side NET-derived profibrotic factors accelerate the extracellular matrix remodeling and contribute to heart valves and/or mesenteric fibrosis development, namely, major complications of functioning NETs. However, at present, little is known on the immune landscape of NETs, but accumulating evidence shows that tumor-infiltrating neutrophils, mast cells, and/or macrophages concur to promote the neoangiogenic switch of these tumors by either direct or indirect mechanisms. On the other hand, immune checkpoint molecules are heterogeneously expressed in NETs' surrounding cells, and it is unclear whether or not tumor-infiltrating lymphocytes are antitumor armed within the microenvironment, given their low mutational load. Here, we review the current knowledge on both gastroenteropancreatic and pulmonary NETs' microenvironment as well as both established and innovative treatments aimed at targeting the tumor-host interplay.


Subject(s)
Neuroendocrine Tumors/pathology , Tumor Microenvironment/physiology , Antineoplastic Agents/classification , Antineoplastic Agents/therapeutic use , Endothelial Cells/physiology , Extracellular Matrix/physiology , Extracellular Traps/physiology , Humans , Immune System/pathology , Immune System/physiology , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology , Neuroendocrine Tumors/blood supply , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/physiopathology , Signal Transduction/physiology , Stromal Cells/physiology
2.
Endocrinol Metab Clin North Am ; 47(3): 471-483, 2018 09.
Article in English | MEDLINE | ID: mdl-30098711

ABSTRACT

Effective management of neuroendocrine tumors depends on early diagnosis, personalized risk stratification, and monitoring response to therapy. During cancer progression, tumors shed circulating tumor cells, circulating tumor DNA, and microRNAs into the bloodstream. Analysis of these biomarkers offers the prospect of a liquid biopsy to predict/monitor therapeutic responses, assess drug resistance, and quantify residual disease. Compared with single-site biopsies, these entities have the potential to inform intratumor heterogeneity and tumor evolution in a reproducible and less invasive way. This article summarizes the state-of-the-art on the potential role of these markers as prognostic and predictive factors in neuroendocrine tumors.


Subject(s)
DNA, Neoplasm/blood , Liquid Biopsy/methods , MicroRNAs/blood , Neoplastic Cells, Circulating/pathology , Neuroendocrine Tumors/blood , Neuroendocrine Tumors/diagnosis , Biomarkers, Tumor/blood , Humans
3.
Oncotarget ; 8(14): 22534-22549, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28186979

ABSTRACT

Neuroendocrine tumors (NETs) metastasize to the skeleton in approximately 20% of patients. We have previously shown that the epithelial-mesenchymal transition (EMT) regulates the NET osteotropism and that CXCR4 overexpression predicts bone spreading. Here, we unravel the molecular mechanisms linking the activation of the CXCL12/CXCR4 axis to the bone colonization of NETs using cell lines representative of pancreatic (BON1, CM, QGP1), intestinal (CNDT 2.5), and bronchial origin (H727). By combining flow cytometry and ELISA, BON1, CM and QGP1 cells were defined as CXCR4high/CXCL12low, while H727 and CNDT 2.5 were CXCR4low/CXCL12high. CXCL12 was inert on cell proliferation, but significantly increased the in vitro osteotropism of CXCR4high/CXCL12low cells, as assessed by transwell assays with or without Matrigel membranes. In these cells, CXCL12 induced in vitro a marked EMT-like transcriptional shift with acquirement of a mesenchymal shape. The nuclei of CXCR4high/CXCL12low NET cells were typically enriched in non-phosphorylated CXCR4, particularly upon agonist stimulation. Silencing of CXCR4 via siRNA prevented the CXCL12-induced EMT in CXCR4high/CXCL12low NET cell lines resulting in the abrogation of both migration and transcriptional mesenchymal patterns. Our data suggest that CXCL12 conveys EMT-promoting signals in NET cells through CXCR4, which in turn regulates transcriptional, morphologic and functional modifications resulting in enhanced in vitro osteotropism of NET cells. Unique functions of CXCR4 may be segregated in relation to its subcellular localization and may acquire potential relevance in future in vivo studies.


Subject(s)
Bone Neoplasms/immunology , Chemokine CXCL12/metabolism , Epithelial-Mesenchymal Transition , Neuroendocrine Tumors/immunology , Receptors, CXCR4/metabolism , Bone Neoplasms/secondary , Bone and Bones/metabolism , Cell Line, Tumor , Cell Movement , Humans , Mesoderm/physiology , Neoplasm Metastasis , Neuroendocrine Tumors/pathology , Phosphorylation , Signal Transduction , Transcriptome , Tropism
4.
Oncotarget ; 7(35): 57414-57429, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27418145

ABSTRACT

High-throughput next-generation sequencing methods have recently provided a detailed picture of the genetic landscape of neuroendocrine tumors (NETs), revealing recurrent mutations of chromatin-remodeling genes and little-to-no pathogenetic role for oncogenes commonly mutated in cancer. Concurrently, multiple epigenetic modifications have been described across the whole spectrum of NETs, and their putative function as tumorigenic drivers has been envisaged. As result, it is still unclear whether or not NETs are epigenetically-driven, rather than genetically-induced malignancies. Although the NET epigenome profiling has led to the identification of molecularly-distinct tumor subsets, validation studies in larger cohorts of patients are needed to translate the use of NET epitypes in clinical practice. In the precision medicine era, recognition of subpopulations of patients more likely to respond to therapeutic agents is critical, and future studies testing epigenetic biomarkers are therefore awaited. Restoration of the aberrant chromatin remodeling machinery is an attractive approach for future treatment of cancer and in several hematological malignancies a few epigenetic agents have been already approved. Although clinical outcomes of epigenetic therapies in NETs have been disappointing so far, further clinical trials are required to investigate the efficacy of these drugs. In this context, given the immune-stimulating effects of epidrugs, combination therapies with immune checkpoint inhibitors should be tested. In this review, we provide an overview of the epigenetic changes in both hereditary and sporadic NETs of the gastroenteropancreatic and bronchial tract, focusing on their diagnostic, prognostic and therapeutic implications.


Subject(s)
Epigenesis, Genetic , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Adaptor Proteins, Signal Transducing/metabolism , Biomarkers, Tumor/genetics , Co-Repressor Proteins , DNA Methylation , Humans , Immune System , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Molecular Chaperones , Mutation , Nuclear Proteins/metabolism , Oncogenes , Prognosis , Proto-Oncogene Proteins/metabolism , X-linked Nuclear Protein/metabolism
5.
Oncologist ; 21(4): 404-17, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26865587

ABSTRACT

UNLABELLED: Obesity is an important risk factor for breast cancer (BC) in postmenopausal women; interlinked molecular mechanisms might be involved in the pathogenesis. Increased levels of estrogens due to aromatization of the adipose tissue, inflammatory cytokines such as tumor necrosis factor-α, interleukin-6, and prostaglandin E2, insulin resistance and hyperactivation of insulin-like growth factors pathways, adipokines, and oxidative stress are all abnormally regulated in obese women and contribute to cancerogenesis. These molecular factors interfere with intracellular signaling in the mitogen-activated protein kinase and phosphatydilinositol-3-phosphate/mammalian target of rapamycin (mTOR) pathways, which regulate the progression of the cell cycle, apoptosis, and protein synthesis. In this context, structural defects of typical genes related to both BC and obesity, such as leptin, leptin receptor, serum paraoxonase/arylesterase 1, the fat mass and obesity-associated gene and melanocortin receptor 4, have been associated with a high or low risk of BC development. The early detection of these gene alterations might be useful as risk predictors in obese women, and targeting these pathways involved in the BC pathogenesis in obese women is a potential therapeutic tool. In particular, mTOR pathway deregulation concurs in both obesity and BC, and inhibition of this might disrupt the molecular interlinks in a similar manner to that of metformin, which exerts definite anticancer activity and is currently used as an antidiabetic drug with a weight-reducing property. The identification of both genetic and pharmacological implications on the prevention and management of BC is the ultimate aim of these studies. IMPLICATIONS FOR PRACTICE: Obese women are at risk of breast cancer, but clinicians lack concrete tools for the prevention or early diagnosis of this risk. The present study, starting from the biology and the molecular defects characterizing both obesity and breast cancer, analyzed the potential molecules and genetic defects whose early identification could delineate a risk profile. Three steps are proposed that are potentially achievable in the clinical assessment of obese women, namely the evaluation of altered levels of serum molecules, the identification of genetic polymorphisms, and the study of the transcriptomic profile of premalignant lesions. Finally, the therapeutic implications of this molecular assessment were evaluated.


Subject(s)
Breast Neoplasms/genetics , Inflammation/genetics , Insulin Resistance/genetics , Obesity/genetics , Breast Neoplasms/etiology , Breast Neoplasms/pathology , Estrogens/genetics , Female , Humans , Inflammation/complications , Inflammation/pathology , Interleukin-6/genetics , Obesity/complications , Obesity/pathology , Postmenopause/genetics , Risk Factors , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/genetics
6.
BMC Cancer ; 16: 87, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26867653

ABSTRACT

BACKGROUND: Gastrointestinal stromal tumors (GISTs) are characterized by mutations of KIT (v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog) or PDGFRA (platelet-derived growth factor receptor α) that may be efficiently targeted by tyrosine kinase inhibitors (TKI). Notwithstanding the early responsiveness to TKI, the majority of GISTs progress, imposing the need for alternative therapeutic strategies. DOG1 (discovered on GIST-1) shows a higher sensitivity as a diagnostic marker than KIT, however its prognostic role has been little investigated. METHODS: We evaluated DOG1 expression by immunohistochemistry (IHC) in 59 patients with GISTs, and correlated its levels with clinical and pathological features as well as mutational status. Kaplan-Meier analysis was also applied to assess correlations of the staining score with patient recurrence-free survival (RFS). RESULTS: DOG1 was expressed in 66% of CD117(+) GISTs and highly associated with tumor size and the rate of wild-type tumors. Kaplan-Meier survival analysis showed that a strong DOG1 expression demonstrated by IHC correlated with a worse 2-year RFS rate, suggesting its potential ability to predict GISTs with poor prognosis. CONCLUSIONS: These findings suggest a prognostic role for DOG1, as well as its potential for inclusion in the criteria for risk stratification.


Subject(s)
Chloride Channels/biosynthesis , Gastrointestinal Stromal Tumors/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Recurrence, Local/genetics , Proto-Oncogene Proteins c-kit/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Adult , Aged , Aged, 80 and over , Anoctamin-1 , Chloride Channels/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Mutation , Neoplasm Proteins/genetics , Neoplasm Recurrence, Local/pathology , Prognosis
7.
Crit Rev Oncol Hematol ; 95(1): 1-11, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25744785

ABSTRACT

Erdheim-Chester disease (ECD) is a rare form of non-Langerhans-cell histiocytosis, associated in more than 50% of cases to BRAF(V600E) mutations in early multipotent myelomonocytic precursors or in tissue-resident histiocytes. It encompasses a spectrum of disorders ranging from asymptomatic bone lesions to multisystemic, life-threatening variants. We reviewed all published reports of histologically-confirmed ECD and explored clinical, radiological, prognostic and therapeutic characteristics in a population of 448 patients, including a unique patient from our Department. To find a clinically relevant signature defining differentiated prognostic profiles, the patients' disease features were compared in relation to their CNS involvement that occurred in 56% of the entire population. Diabetes insipidus, visual disturbances, pyramidal and extra-pyramidal syndromes were the most recurrent neurological signs, whereas concomitant pituitary involvement, retro-orbital masses and axial lesions in the presence of symmetric bilateral osteosclerosis of long bones depicted the typical ECD clinical picture. Patients with CNS infiltration showed a lower occurrence of heart involvement and a higher incidence of bone, skin, retro-peritoneal, lung, aortic and renal infiltration. No difference in the therapeutic algorithm was found after stratification for CNS involvement. A better understanding of the disease pathogenesis, including BRAF deregulation, in keeping with improved prognostic criteria, will provide novel suggestions for the management of ECD.


Subject(s)
Erdheim-Chester Disease/pathology , Adrenal Cortex Hormones/therapeutic use , Adult , Bone and Bones/pathology , Central Nervous System/pathology , Erdheim-Chester Disease/diagnosis , Erdheim-Chester Disease/drug therapy , Erdheim-Chester Disease/genetics , Humans , Kidney/pathology , Male , Myocardium/pathology , Point Mutation , Prognosis , Proto-Oncogene Proteins B-raf/genetics
8.
Cell Signal ; 25(5): 1108-17, 2013 May.
Article in English | MEDLINE | ID: mdl-23380051

ABSTRACT

Melphalan has been a mainstay of multiple myeloma (MM) therapy for many years. However, following treatment with this alkylator, malignant plasma cells usually escape both apoptosis and cell cycle control, and acquire drug-resistance resulting in tumor progression. Bendamustine is being used in MM patients refractory to conventional DNA-damaging agents, although the mechanisms driving this lack of cross-resistance are still undefined. Here, we investigated the molecular pathway of bendamustine-induced cell death in melphalan-sensitive and melphalan-resistant MM cell lines. Bendamustine affected cell survival resulting in secondary necrosis, and prompted cell death primarily through caspase-2 activation. Also, bendamustine blocked the cell cycle in the G2/M phase and induced micronucleation, erratic chromosome spreading and mitotic spindle perturbations in melphalan-resistant MM cells. In these cells, both Aurora kinase A (AURKA) and Polo-like kinase-1 (PLK-1), key components of the spindle-assembly checkpoint, were down-regulated following incubation with bendamustine, whereas levels of Cyclin B1 increased as a consequence of the prolonged mitotic arrest induced by the drug. These findings indicate that, at least in vitro, bendamustine drives cell death by promoting mitotic catastrophe in melphalan-resistant MM cells. Hence, activation of this alternative pathway of cell death may be a novel approach to the treatment of apoptosis-resistant myelomas.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Melphalan/pharmacology , Mitosis , Nitrogen Mustard Compounds/pharmacology , Aurora Kinase A , Aurora Kinases , Bendamustine Hydrochloride , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cyclin B1/metabolism , Drug Resistance, Neoplasm/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...