Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
J Med Vasc ; 43(4): 246-254, 2018 Jul.
Artigo em Francês | MEDLINE | ID: mdl-29981733

RESUMO

Pancreatic cancer (PC) is a devastating malignancy with an overall 5-year survival of 8% for all stages combined. Most of the PC patients diagnosed have an advanced disease (40%) or metastatic stage (40%), which eliminates surgery as a potentially curative treatment. The disease course is often complicated by venous thromboembolism (VTE) events, which per se account for significant morbidity and mortality, with significantly worsen survival. PC is associated with the highest risk of VTE among all cancer patients. We review the literature data to address the incidence and clinical outcomes of VTE in PC patients. VTE incidence varies from 5 to 41% according to epidemiological studies and is as high as 57% in postmortem series. Since 2013, international clinical practice guidelines recommend primary thromboprophylaxis with a grade 1B level of evidence as an adjuvant therapy in advanced PC. A recent meta-analysis of randomized controlled trials investigating the benefit and risk of low-molecular-weight heparins (LMWH) in ambulatory advanced PC patients under chemotherapy showed that the incidence of VTE was 2.1% in patients treated with LMWH and 11.2% in controls (risk ratio, 0.18; 95% CI, 0.083-0.39; P<0.0001). In conclusion, improved earlier diagnosis and effective management of VTE, a frequent and life-threatening complication in PC, is warranted to improve PC patient outcomes.


Assuntos
Neoplasias Pancreáticas/complicações , Tromboembolia Venosa/etiologia , Anticoagulantes/uso terapêutico , Diagnóstico Precoce , Heparina de Baixo Peso Molecular/uso terapêutico , Humanos , Incidência , Metanálise como Assunto , Neoplasias Pancreáticas/sangue , Síndrome Pós-Trombótica/etiologia , Guias de Prática Clínica como Assunto , Prevalência , Estudos Retrospectivos , Taxa de Sobrevida , Trombofilia/tratamento farmacológico , Trombofilia/etiologia , Trombofilia/fisiopatologia , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/epidemiologia , Tromboembolia Venosa/prevenção & controle
2.
Colorectal Dis ; 17(4): 311-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25425534

RESUMO

AIM: Combined infliximab and sphincter-sparing surgery can be effective in perianal fistula associated with Crohn's disease (CD). This study aimed to assess the efficacy of local surgery combined with infliximab on sustained fistula closure and to identify predictive factors for response after this combined treatment. METHOD: Between 2000 and 2010, 81 patients with fistulising perianal CD were included in this observational study. Drainage with a loose seton was followed by infliximab therapy. The primary end-points were the rate of complete fistula closure and time required for this to occur. RESULTS: The fistula was complex in 71 (88%) of the 81 patients. Local proctological surgery was carried out in 77 (95%), including seton drainage in 62 (80.5%) of these. This was continued for a median duration of 3.8 months and the patient then received infliximab therapy. The median follow-up after treatment was 64 months (2-263). Initial complete closure of the fistula occurred in 71 (88%) cases at a median interval of 12.4 months (1-147) from the start of treatment. Recurrence was observed in 29 (41%) patients at a median interval of 38.5 months (2-48) from the start of treatment. They were treated again with combined treatment with successful closure in 19 (65.5%) patients. The total rate of closure of the fistula was 75.3%. Female gender, anal stenosis, rectovaginal and complex fistula formation were factors independently associated with failure of combined treatment. CONCLUSION: Seton drainage for several months combined with infliximab therapy is effective in closing the fistula in 75% of patients with complex perianal fistula formation associated with CD.


Assuntos
Doença de Crohn/terapia , Fármacos Gastrointestinais/uso terapêutico , Infliximab/uso terapêutico , Fístula Retal/terapia , Adolescente , Adulto , Canal Anal , Estudos de Coortes , Terapia Combinada , Doença de Crohn/complicações , Drenagem/métodos , Feminino , Humanos , Masculino , Tratamentos com Preservação do Órgão , Fístula Retal/etiologia , Estudos Retrospectivos , Resultado do Tratamento , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adulto Jovem
3.
Pancreatology ; 12(1): 27-34, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22487470

RESUMO

AIMS: The purpose of this study was to investigate the clinical feasibility and utility of low-density array analysis on samples obtained from endoscopic ultrasound-guided fine needle aspiration biopsy in locally advanced and/or metastatic pancreatic ductal adenocarcinoma and chronic pancreatitis. PATIENTS AND METHODS: In this prospective multicenter study, we quantified candidate gene expression in biopsies sampled from 44 locally advanced and/or metastatic pancreatic carcinoma and from 17 pseudotumoural chronic pancreatitis using dedicated low-density array microfluidic plates. RESULTS: We first demonstrated that 18S gene expression is stable and comparable in normal pancreas and pancreatic cancer tissues. Next, we found that eight genes (S100P, PLAT, PLAU, MSLN, MMP-11, MMP-7, KRT7, KRT17) were significantly over expressed in pancreatic cancer samples when compared to pseudotumoural chronic pancreatitis (p value ranging from 0.0007 to 0.0215): Linear discriminative analysis identified S100P, PLAT, MSLN, MMP-7, KRT7 as highly explicative variables. The area under receiver operating curve establishes the clinical validity of the potential diagnostic markers identified in this study (values ranging from 0.69 to 0.76). In addition, combination of S100P and KRT7 gave better diagnosis performances (Area Under Receiver Operating Curve 0.81, sensitivity 81%, specificity 77%). CONCLUSION: We demonstrate that molecular studies on EUS-guided FNA material are feasible for the identification and quantification of markers in PDAC patients diagnosed with non-resectable tumours. Using low-density array, we isolated a molecular signature of advanced pancreatic carcinoma including mostly cancer invasion-related genes. This work stems for the use of novel biomarkers for the molecular diagnosis of patient with solid pancreatic masses.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Biópsia por Agulha Fina , Carcinoma Ductal Pancreático/diagnóstico por imagem , Carcinoma Ductal Pancreático/patologia , Endossonografia , Perfilação da Expressão Gênica , Humanos , Mesotelina , Pâncreas/diagnóstico por imagem , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/patologia , Pancreatite Crônica , Estudos Prospectivos , Sensibilidade e Especificidade
4.
J Oncol ; 2010: 178174, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21052499

RESUMO

Pancreatic cancer is the eighth most common cause of death from cancer in the world, for which palliative treatments are not effective and frequently accompanied by severe side effects. We propose a DNA-based therapy for pancreatic cancer using a nonviral vector, expressing the diphtheria toxin A chain under the control of the H19 gene regulatory sequences. The H19 gene is an oncofetal RNA expressed during embryo development and in several types of cancer. We tested the expression of H19 gene in patients, and found that 65% of human pancreatic tumors analyzed showed moderated to strong expression of the gene. In vitro experiments showed that the vector was effective in reducing Luciferase protein activity on pancreatic carcinoma cell lines. In vivo experiment results revealed tumor growth arrest in different animal models for pancreatic cancer. Differences in tumor size between control and treated groups reached a 75% in the heterotopic model (P = .037) and 50% in the orthotopic model (P = .007). In addition, no visible metastases were found in the treated group of the orthotopic model. These results indicate that the treatment with the vector DTA-H19 might be a viable new therapeutic option for patients with unresectable pancreatic cancer.

5.
Cancer Gene Ther ; 17(5): 315-24, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19911032

RESUMO

Pancreatic cancer (PC) remains a life-threatening disease. Efficient therapeutic gene delivery to PC-derived cells continues to present challenges. We used self-inactivated lentiviral vectors to transduce PC-derived cells in vitro and in vivo. We showed that lentiviral vectors transduce PC-derived cell lines with high efficiency (>90%), regardless of the differentiation state of the cell. Next, we transferred human interferon beta (hIFN-beta) gene. Expression of hIFN-beta in PC cells using lentiviral vectors resulted in the inhibition of cell proliferation and the induction of cell death by apoptosis. In vivo, lentiviral administration of hIFN-beta prevented PC tumor progression for up to 15 days following gene therapy, and induced tumor regression/stabilization in 50% of the mice treated. Again, hIFN-beta expression resulted in cancer cell proliferation inhibition and apoptosis induction. We provide evidence that human immunodeficiency virus (HIV)-1-based lentiviral vectors are very efficient for gene transfer in PC-derived cells in vitro and in vivo. As a consequence, delivery of hIFN-beta stopped PC tumor progression. Thus, our approach could be applied to the 85% of PC patients with a locally advanced disease.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/genética , Lentivirus/genética , Neoplasias Pancreáticas/terapia , Animais , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Interferon beta/genética , Interferon beta/fisiologia , Camundongos , Camundongos SCID , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Endoscopy ; 41(6): 552-7, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19533561

RESUMO

BACKGROUND AND STUDY AIMS: Differential diagnosis between pancreatic adenocarcinoma (PADC) and pseudotumoral forms of chronic pancreatitis remains difficult. Mutation of KRAS oncogene is present in 75% to 95% of PADC. This study aimed to evaluate whether the combined analysis of KRAS mutation with cytopathological findings from endoscopic ultrasound-guided fine-needle aspiration biopsy (EUS-FNAB) might improve discrimination between PADC and chronic pancreatitis. PATIENTS AND METHODS: This prospective multicenter study included 178 patients with solid pancreatic masses (men 104, women 74; mean age 64.5 years). Cytopathological examination and KRAS mutation analysis (codon-12 and codon-13, restriction fragment length polymorphism [RFLP] and direct sequencing) were performed on EUS-FNAB material. Final diagnoses were obtained on EUS-FNAB analysis and/or a second biopsy and/or clinical follow-up and/or surgery: PADC, n = 129; chronic pancreatitis, n = 27; other pancreatic neoplasms, n = 16; and benign lesions, n = 6. RESULTS: KRAS status analysis was successful in all EUS-FNAB samples. Codon-12 KRAS point mutation was found in 66% of PADC samples. No case of chronic pancreatitis displayed KRAS mutation. Sensitivity, specificity, positive and negative predictive values, and overall accuracy of cytopathology alone for diagnosis of PADC versus chronic pancreatitis were 83%, 100%, 100%, 56% and 86%, respectively. When KRAS mutation analysis was combined with cytopathology, these values reached 88%, 100%, 100%, 63% and 90% respectively. CONCLUSION: Although the value of KRAS analysis in addition to EUS-FNAB is limited for distinguishing pancreatic mass lesions, when chronic pancreatitis presented as a pseudotumor a negative finding (wild-type KRAS), was useful in strongly suggesting a benign lesion.


Assuntos
Endossonografia , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Pancreatite Crônica/patologia , Idoso , Biópsia por Agulha Fina , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/genética , Pancreatite Crônica/diagnóstico por imagem , Pancreatite Crônica/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Proteínas ras/genética
9.
Cell Death Differ ; 14(6): 1202-10, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17347668

RESUMO

Hepatocellular carcinoma (HCC) is a major public health concern because of the absence of early diagnosis and effective treatments. Efficient diagnosis modalities and therapies to treat HCC are needed. Kruppel-like factor (KLF) family members, such as KLF6, are involved in cell proliferation and differentiation. KLF6 is inactivated in solid tumors, which may contribute to pathogenesis. However, KLF6 status in HCC is controversial. Thus, we undertook the characterization of KLF6 expression and function in HCC and HCC-derived cell lines. We found that HCC, HepG2 and HuH7 cells expressed KLF6 messenger ribonucleic acid and protein. Next, using RNA interference, we demonstrated that inhibiting KLF6 expression in vitro strongly impaired cell proliferation-induced G1-phase arrest, inhibited cyclin-dependent kinase 4 and cyclin D1 expression, and subsequent retinoblastoma phosphorylation. Finally, KLF6 silencing caused p53 upregulation and inhibited Bcl-xL expression, to induce cell death by apoptosis. Taken together, these data demonstrated that KLF6 is essential for HCC-derived cells to evade apoptosis.


Assuntos
Apoptose/fisiologia , Fatores de Transcrição Kruppel-Like/fisiologia , Apoptose/genética , Sequência de Bases , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Ciclo Celular/genética , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Dados de Sequência Molecular , Mutação , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/genética , Proteína do Retinoblastoma/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico
10.
Cell Death Differ ; 14(2): 197-208, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16645635

RESUMO

Somatostatin is a multifunctional hormone that modulates cell proliferation, differentiation and apoptosis. Mechanisms for somatostatin-induced apoptosis are at present mostly unsolved. Therefore, we investigated whether somatostatin receptor subtype 2 (sst2) induces apoptosis in the nontransformed murine fibroblastic NIH3T3 cells. Somatostatin receptor subtype 2 expression induced an executioner caspase-mediated apoptosis through a tyrosine phosphatase SHP-1 (Src homology domain phosphatase-1)-dependent stimulation of nuclear factor kappa B (NF-kappaB) activity and subsequent inhibition of the mitogen-activated protein kinase JNK. Tumor necrosis factor alpha (TNFalpha) stimulated both NF-kappaB and c-Jun NH2-terminal kinase (JNK) activities, which had opposite action on cell survival. Importantly, sst2 sensitized NIH3T3 cells to TNFalpha-induced apoptosis by (1) upregulating TNFalpha receptor protein expression, and sensitizing to TNFalpha-induced caspase-8 activation; (2) enhancing TNFalpha-mediated activation of NF-kappaB, resulting in JNK inhibition and subsequent executioner caspase activation and cell death. We have here unraveled a novel signaling mechanism for a G protein-coupled receptor, which directly triggers apoptosis and crosstalks with a death receptor to enhance death ligand-induced apoptosis.


Assuntos
Apoptose , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Receptores de Somatostatina/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Somatostatina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Camundongos , Modelos Biológicos , Mimetismo Molecular/efeitos dos fármacos , Células NIH 3T3 , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 1 , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais/efeitos dos fármacos , Transfecção , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia
11.
Cancer Gene Ther ; 14(1): 19-29, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16990845

RESUMO

Pancreatic cancer is one of the most aggressive and devastating human malignancies. There is an urgent need for more effective therapy for patients with advanced disease. In this context, genetic therapy potentially represents a rational new approach to treating pancreatic cancer, which could provide an adjunct to conventional options. Because of the promise of recombinant SV40 vectors, we tested their ability to deliver a transgene, and to target a transcript, so as to inhibit pancreatic tumors growth in vivo. BxPC3 and Capan-1 cells were efficiently transduced using SV40 vectors without selection, as compared to synthetic vectors PEI. SV40 vectors were as efficient as adenoviral vectors, and provided long-term transgene expression. Next, we devised a SV40-derived, targeted gene therapy approach of pancreatic cancer, by combining hTR tumor-specific promoter with sst2 somatostatin receptor tumor-suppressor gene. In vitro cell proliferation was strongly impaired following administration of SV(hTR-sst2). SV40-derived sst2-mediated antiproliferative effect was dependent on the local production of somatostatin. In vivo, intratumoral gene transfer of sst2 using rSV40 vectors resulted in a marked inhibition of Capan-1 tumor progression, and proliferation. These results represent the initial steps toward a novel approach to the gene therapy of pancreatic cancer using SV40 as a vector.


Assuntos
Vírus Defeituosos/fisiologia , Técnicas de Transferência de Genes , Neoplasias Pancreáticas/patologia , Vírus 40 dos Símios/fisiologia , Replicação Viral , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Humanos , Reação em Cadeia da Polimerase , Transdução Genética
12.
Gut ; 56(1): 107-14, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16891358

RESUMO

BACKGROUND: Peritoneal carcinomatosis from pancreatic cancer has a poor prognosis with a median survival of 3.1 months. This is mainly due to lack of effective treatment. Interleukin 12 (IL12) is a proinflammatory cytokine that has a potent antitumoral effect by stimulating innate and adoptive immunity. AIM: To examine the antitumoral effect and toxicity of intraperitoneal delivery of IL12 using an ex vivo gene therapy approach in a murine model of pancreatic peritoneal carcinomatosis. METHODS: Peritoneal carcinomatosis was generated by direct intraperitoneal inoculation of the pancreatic cancer cell line Capan-1 in athymic mice. Syngenic fibroblasts were genetically modified in vitro to secrete IL12 using a polycistronic TFG murine IL12 retroviral vector coding for both p35 and p40 murine IL12 subunits. Ex vivo gene therapy involved injection of the genetically modified fibroblasts intraperitoneally twice a week for 4 weeks. RESULTS: Treatment of pre-established peritoneal carcinomatosis with fibroblasts genetically modified to express IL12 induced a marked inhibition of tumour growth as measured by comparison of the weights of the intraperitoneal tumour nodules in the treated and control animals (3.52 (SD 0.47) v 0.93 (SD 0.21) g, p<0.05) and improved survival. This effect was associated with infiltration of the peritoneal tumour nodules with macrophages. Peritoneal lavage confirmed enhancement of the innate peritoneal inflammatory activity, with an increased number of activated macrophages and natural killer cells. Moreover, macrophages harvested from animals with peritoneal carcinomatosis and treated with IL12-expressing fibroblasts expressed an activated proinflammatory antitumoral M1 phenotype that included strongly enhanced reactive oxygen species and nitric oxide production. There was no treatment-related toxicity. CONCLUSION: Multiple injections of genetically modified fibroblasts to express IL12 is an effective and well-tolerated treatment for experimental murine pancreatic peritoneal carcinomatosis via activated innate immunity and in particular activated M1 macrophages.


Assuntos
Antineoplásicos/imunologia , Fibroblastos/imunologia , Terapia Genética/métodos , Interleucina-12/imunologia , Neoplasias Peritoneais/terapia , Animais , Divisão Celular/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Citometria de Fluxo/métodos , Imunidade Inata/imunologia , Imuno-Histoquímica/métodos , Injeções Intraperitoneais , Interleucina-12/administração & dosagem , Interleucina-12/genética , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Óxido Nítrico/biossíntese , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/imunologia , Espécies Reativas de Oxigênio/metabolismo
13.
Ann Oncol ; 17(12): 1733-42, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16801334

RESUMO

BACKGROUND: There is a need for novel antitumor agents that demonstrate efficacy in currently refractory tumors without adding to the toxicity of therapy. The somatostatin analogs, which have demonstrated antineoplastic activities in experimental tumor models, and good tolerability and safety profiles are attractive candidates. MATERIALS AND METHODS: Data from preclinical studies provide evidence for direct and indirect mechanisms by which somatostatin analogs exert antitumor effects. RESULTS: Direct antitumor activities, mediated through somatostatin receptors (sst(1)-sst(5)) expressed in tumor cells, include blockade of autocrine/paracrine growth-promoting hormone and growth factor production, inhibition of growth factor-mediated mitogenic signals and induction of apoptosis. Indirect antitumor effects include inhibition of growth-promoting hormone and growth factor secretion, and antiangiogenic actions. Many human tumors express more than one somatostatin receptor subtype, with sst(2) being predominant. Somatostatin analogs such as octreotide and lanreotide, which present a high affinity for sst(2), are in current clinical use to alleviate symptoms in patients with endocrine tumors, and radiolabeled somatostatin analogs have been developed for diagnosis and radiotherapy. CONCLUSIONS: While the rationale exists for the use of somatostatin analogs as antitumor agents, studies are ongoing to identify analogs with activity across the range of receptor subtypes to maximize the potential of such treatment.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Somatostatina/análogos & derivados , Humanos , Somatostatina/uso terapêutico
14.
Endoscopy ; 38(4): 349-54, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16680633

RESUMO

BACKGROUND AND STUDY AIM: Endoscopic ultrasonography (EUS) now has an important place in the diagnosis of gastroenteropancreatic diseases. However, prospective data on the morbidity and mortality related to its use are sparse and often retrospective. We attempted to assess the acute and immediate complications of both diagnostic and interventional EUS. PATIENTS AND METHODS: At our university-affiliated tertiary care referral center, immediate (occurring during the procedure) and acute (occurring within 24 hours) complications of EUS were prospectively investigated. Over a first period, spanning 10 years, complications of diagnostic EUS involving 3207 consecutive patients were assessed. During the second period of 3 years, complications observed after EUS-guided fine-needle aspiration (FNA) biopsy were evaluated from 224 procedures. EUS was mostly done with the patient under sedation with intravenous propofol and spontaneous ventilation, and complications were evaluated by both the operator and the anesthesiologist. RESULTS: There were no deaths, and no surgery was required over the two periods of assessment. Three mild complications occurred among patients who underwent standard diagnostic EUS: two immediate complications were related to anesthesia and one to the procedure. There were five complications associated with interventional EUS; all were related to the procedure (acute pancreatitis, duodenal perforation, upper digestive bleeding, cyst, and mediastinal infection), with a mean delay of occurrence of 30 hours, and mean duration of hospitalization of 7 days. CONCLUSION: In our experience, which is the longest reported in Europe, the morbidity rates of diagnostic EUS and EUS-guided FNA biopsy were 0.093% and 2.2%, respectively, with no mortality.


Assuntos
Duodeno/lesões , Endossonografia/efeitos adversos , Endossonografia/mortalidade , Hemorragia Gastrointestinal/etiologia , Neoplasias Pancreáticas/diagnóstico por imagem , Encaminhamento e Consulta , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia por Agulha Fina , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/patologia , Estudos Prospectivos , Ruptura , Taxa de Sobrevida
15.
Dig Liver Dis ; 36 Suppl 1: S2-7, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15077905

RESUMO

Somatostatin is an inhibitory neuropeptide, which acts on various targets throughout the body to regulate a variety of physiological functions including inhibition of endocrine and exocrine secretions, modulation of neurotransmission, motor and cognitive functions, inhibition of intestinal motility, absorption of nutrients and ions, vascular contractility and inhibition of normal and tumour cell proliferation. It exerts its effects through interaction with five somatostatin receptors (sst1-sst5), which belong to the family of G-protein-coupled receptors with seven transmembrane spanning domains and are variably expressed in a variety of tumours such as gastroenteropancreatic tumours, pituitary tumours, and carcinoid tumours. This review covers the present knowledge regarding the molecular mechanisms involved in somatostatin antineoplastic activity. Evidence that sst2 receptor acts as a tumour suppressor is also discussed.


Assuntos
Divisão Celular/fisiologia , Receptores de Somatostatina/fisiologia , Somatostatina/fisiologia , Proteínas Ativadoras de GTPase/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia
16.
Ann Endocrinol (Paris) ; 63(2 Pt 3): 2S13-8, 2002 Apr.
Artigo em Francês | MEDLINE | ID: mdl-12037498

RESUMO

Somatostatin and its stable analogues (octreotide, lanreotide and vapreotide) exert an antiproliferative effect on various normal and cancerous cells both in vitro and in vivo. This effect results from different mechanisms: an indirect effect by the inhibition of release of growth factors and trophic hormones (GH, IGF-1, insulin, gastrin, EGF), an inhibition of angiogenesis processes (endothelial cell proliferation, VEGF release, monocyte activity), an immunomodulatory effect (lymphocyte proliferation, interleukine or cytokine release, NK activity) and a direct effect on target cells. This direct antiproliferative effect is mediated through specific somatostatin receptors. Among them, sst(1), sst(2), sst(4) and sst(5) have been implicated in vitro in the G1-G0 cell cycle blockade, sst(3) and sst(2) mediating the apoptotic effect of somatostatin. In addition, sst(2) acts as an antioncogene in human pancreatic cancer cells. Coupling to membrane tyrosine phosphatases (SHP-1, SHP-2) is the main transduction pathway involved in the antiproliferative effect mediated by sst receptors. The dissociation observed clinically between a frequent antisecretory response and an inconstant antitumor effect after administration of somatostatin analogues may reflect an absence of expression or coupling of the receptor(s) involved in antiproliferative effect. Moreover, a desensitization or mutation of these receptors may also occur in tumors. All the potential mechanism involved should be elucidated in order to improve or better target the antitumor effect of somatostatin analogues clinically used.


Assuntos
Antagonistas de Hormônios/farmacologia , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Humanos , Receptores de Somatostatina/efeitos dos fármacos
17.
FASEB J ; 15(12): 2300-2, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11511520

RESUMO

Somatostatin receptor sst2 is an inhibitory G protein-coupled receptor, which inhibits normal and tumor cell growth by a mechanism involving the tyrosine phosphatase SHP-1. We reported previously that SHP-1 associates transiently with and is activated by sst2 and is a critical component for sst2 growth inhibitory signaling. Here, we demonstrate that in Chinese hamster ovary cells expressing sst2, SHP-1 is associated at the basal level with the neuronal nitric oxide synthase (nNOS). Following sst2 activation by the somatostatin analog RC-160, SHP-1 rapidly recruits nNOS tyrosine dephosphorylates and activates it. The resulting NO activates guanylate cyclase and inhibits cell proliferation. Coexpression of a catalytically inactive SHP-1 mutant with sst2 blocks RC-160-induced nNOS dephosphorylation and activation, as well as guanylate cyclase activation. In mouse pancreatic acini, RC-160 treatment reduces nNOS tyrosine phosphorylation accompanied by an increase of its activity. By opposition, in acini from viable motheaten (mev/mev) mice, which express a markedly inactive SHP-1, RC-160 has no effect on nNOS activity. Finally, expression of a dominant-negative form of nNOS prevents both RC-160-induced p27 up-regulation and cell proliferation inhibition. We therefore identified nNOS as a novel SHP-1 substrate critical for sst2-induced cell-growth arrest.


Assuntos
Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase/fisiologia , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Somatostatina/fisiologia , Transdução de Sinais , Animais , Células CHO , Divisão Celular , Cricetinae , GMP Cíclico/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Modelos Biológicos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo I , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/fisiologia , Somatostatina/farmacologia
18.
Presse Med ; 30(18): 921-9, 2001 May 19.
Artigo em Francês | MEDLINE | ID: mdl-11413855

RESUMO

NATURAL HISTORY: Alcoholism is a common cause of chronic calcifying pancreatitis. Abdominal pain, weight loss and acute complications are the early manifestations. Besides chronic pain, about 80% of the patients develop acute pancreatitis, pseudocysts, compression of the main bile duct, serous effusion or digestive bleeding during the first five years of the disease course. Painful episodes are more frequent after 5 to 10 years, with a risk of jaundice and persistent pseudocyst formation. CHRONIC COURSE: Beyond 10 years, calcified stones and fibrosis progressively replace the pancreas parenchyma. Painful events become more parse and after 15 years, diabetes and exocrine insufficiency predominate. TREATMENT: Abstention from alcoholic beverages, analgesics administration and balanced diet are essential therapeutic elements. Insulin is required for the diabetes and pancreas extracts in gastroprotected microcapsules are needed to compensate for the chronic exocrine insufficiency. This medical treatment may be able to control pain but interventional radiology (pseudocyst drainage, antalgesic infiltration of the celiac plexus), interventional endoscopy (pseudocyst drainage, biliary stent, pancreatic endoprosthesis), or surgery (deviation of the bile ducts, wirsungojejunal derivation, exceptionally resection) may also be required.


Assuntos
Pancreatite/diagnóstico , Doença Crônica , Terapia Combinada , Diagnóstico Diferencial , Fibrose , Humanos , Pâncreas/patologia , Testes de Função Pancreática , Pancreatite/etiologia , Pancreatite/terapia , Prognóstico
19.
J Biol Chem ; 276(16): 13461-8, 2001 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-11278805

RESUMO

The sst2 somatostatin receptor is an inhibitory G protein-coupled receptor, which exhibits anti-tumor properties. Expression of sst2 is lost in most human pancreatic cancers. We have cloned 2090 base pairs corresponding to the genomic DNA region upstream of the mouse sst2 (msst2) translation initiation codon (ATG). Deletion reporter analyses in mouse pituitary AtT-20 and human pancreatic cancer PANC-1, BxPC-3, and Capan-1 cells identify a region from nucleotide -260 to the ATG codon (325 base pairs) showing maximal activity, and a region between nucleotides -2025 and -260 likely to comprise silencer or transcriptional suppressor elements. In PANC-1 and AtT-20 cells, transforming growth factor (TGF)-beta up-regulates msst2 transcription. Transactivation is mediated by Smad4 and Smad3. The cis-acting region responsible for such regulation is comprised between nucleotides -1115 and -972 and includes Sp1 and CAGA-box sequences. Expression of Smad4 in Smad4-deficient Capan-1 and BxPC-3 cells restores TGF-beta-dependent and -independent msst2 transactivation. Expression of Smad4 in BxPC-3 cells reestablishes both endogenous sst2 expression and somatostatin-mediated inhibition of cell growth. These findings demonstrate that msst2 is a new target gene for TGF-beta transcription regulation and underlie the possibility that loss of Smad4 contributes to the lack of sst2 expression in human pancreatic cancer, which in turn may contribute to a stimulation of tumor growth.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Regiões Promotoras Genéticas , Receptores de Somatostatina/genética , Transativadores/metabolismo , Ativação Transcricional , Fator de Crescimento Transformador beta/farmacologia , Regiões 5' não Traduzidas/genética , Animais , Sequência de Bases , Clonagem Molecular , Códon , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Biblioteca Genômica , Humanos , Luciferases/genética , Camundongos , Dados de Sequência Molecular , Neoplasias Pancreáticas , Neoplasias Hipofisárias , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deleção de Sequência , Proteína Smad4 , Transcrição Gênica , Ativação Transcricional/efeitos dos fármacos , Células Tumorais Cultivadas
20.
Chemotherapy ; 47 Suppl 2: 30-9, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11275700

RESUMO

Over the past decade, antiproliferative effects of somatostatin and analogs have been reported in many somatostatin receptor-positive normal and tumor cell types. Regarding the molecular mechanisms involved, somatostatin or analogs mediate their action through both indirect and direct effects. Somatostatin acts through five somatostatin receptors (SSTR1-5) which are variably expressed in normal and tumor cells. These receptors regulate a variety of signal transduction pathways including inhibition of adenylate cyclase, regulation of ion channels, regulation of serine/threonine and tyrosine kinases and phosphatases. This review focuses on recent advances in biological mechanisms involved in the antineoplastic activity of somatostatin and analogs.


Assuntos
Antineoplásicos Hormonais/farmacologia , Divisão Celular/efeitos dos fármacos , Somatostatina/farmacologia , Animais , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Receptores de Somatostatina/metabolismo , Transdução de Sinais , Somatostatina/análogos & derivados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...