Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EJNMMI Res ; 11(1): 115, 2021 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-34727262

RESUMO

Recent advancements in the field of immune-oncology have led to a significant increase in life expectancy of patients with diverse forms of cancer, such as hematologic malignancies, melanoma and lung cancer. Unfortunately, these encouraging results are not observed in the majority of patients, who remain unresponsive and/or encounter adverse events. Currently, researchers are collecting more insight into the cellular and molecular mechanisms that underlie these variable responses. As an example, the human lymphocyte activation gene-3 (huLAG-3), an inhibitory immune checkpoint receptor, is increasingly studied as a therapeutic target in immune-oncology. Noninvasive molecular imaging of the immune checkpoint programmed death protein-1 (PD-1) or its ligand PD-L1 has shown its value as a strategy to guide and monitor PD-1/PD-L1-targeted immune checkpoint therapy. Yet, radiotracers that allow dynamic, whole body imaging of huLAG-3 expression are not yet described. We here developed single-domain antibodies (sdAbs) that bind huLAG-3 and showed that these sdAbs can image huLAG-3 in tumors, therefore representing promising tools for further development into clinically applicable radiotracers.

2.
Gene Ther ; 24(6): 370-375, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28540936

RESUMO

Targeting transgene expression specifically to antigen-presenting cells (APCs) has been put forward as a promising strategy to direct the immune system towards immunity. We developed the nanobody-display technology to restrict the tropism of lentiviral vectors (LVs) to APCs. However, we observed that immunization with APC-targeted LVs (DC2.1-LVs) did not evoke strong antigen-specific T-cell immunity when compared to immunization with broad tropism LVs (VSV.G-LVs). In this study, we report that VSV.G-LVs are more immunogenic than DC2.1-LVs because they transduce stromal cells, which has a role in activating antigen-specific T cells. Moreover, VSV.G-LVs trigger a pro-inflammatory innate immune response through transduction of APCs and stromal cells, while DC2.1-LVs trigger a type I interferon response with anti-viral capacity. These findings question the rationale of targeting LVs to APCs and argue for the development of VSV.G-LVs with an improved safety profile.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Técnicas de Transferência de Genes/efeitos adversos , Vetores Genéticos/genética , Lentivirus/genética , Linfócitos T/imunologia , Células 3T3 , Animais , Feminino , Terapia Genética/métodos , Vetores Genéticos/efeitos adversos , Células HEK293 , Humanos , Imunidade Inata , Interferon Tipo I/sangue , Camundongos , Camundongos Endogâmicos C57BL
3.
Oncogene ; 34(13): 1650-7, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24793791

RESUMO

The signal transducer and activator of transcription 3 (STAT3) can be activated by the tyrosine kinase domain of the chimeric protein nucleophosmin/anaplastic lymphoma kinase (NPM/ALK), and has a pivotal role in mediating NPM/ALK-related malignant cell transformation. Although the role of STAT3 and wild-type NPM in oncogenesis has been extensively investigated, the relationship between both molecules in cancer remains poorly understood. In the present study, we first demonstrate that STAT3 phosphorylation at tyrosine 705 is accompanied by a concomitant increase in the expression level of NPM. Nuclear co-translocation of phosphorylated STAT3 with NPM can be triggered by interferon-alpha (IFN-α) stimulation of Jurkat cells and phosphorylated STAT3 co-localizes with NPM in cancer cells showing constitutive STAT3 activation. We further demonstrate that STAT3 phosphorylation can transcriptionally mediate NPM upregulation in IFN-α-stimulated Jurkat cells and is responsible for maintaining its expression in cancer cells showing constitutive STAT3 activation. Inhibition of STAT3 phosphorylation or knockdown of NPM expression abrogates their simultaneous transnuclear movements. Finally, we found evidence for a physical interaction between NPM and STAT3 in conditions of STAT3 activation. In conclusion, NPM is a downstream effector of the STAT3 signaling, and can facilitate the nuclear entry of phosphorylated STAT3. These observations might open novel opportunities for targeting the STAT3 pathway in cancer.


Assuntos
Neoplasias/metabolismo , Proteínas Nucleares/fisiologia , Fator de Transcrição STAT3/metabolismo , Transcrição Gênica , Linhagem Celular Tumoral , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Interferon-alfa/farmacologia , Proteínas Nucleares/genética , Nucleofosmina , Fosforilação , Transporte Proteico , Transdução de Sinais/fisiologia
4.
Gene Ther ; 21(3): 262-71, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24401835

RESUMO

The release of cytokines by T cells strongly defines their functional activity in vivo. The ability to produce multiple cytokines has been associated with beneficial immune responses in cancer and infectious diseases, while their progressive loss is associated with T-cell exhaustion, senescence and anergy. Consequently, strategies that enhance the multifunctional status of T cells are a key for immunotherapy. Dendritic cells (DCs) are professional antigen presenting cells that regulate T-cell functions by providing positive and negative co-stimulatory signals. A key negative regulator of T-cell activity is provided by binding of programmed death-1 (PD-1) receptor on activated T cells, to its ligand PD-L1, expressed on DCs. We investigated the impact of interfering with PD-L1/PD-1 co-stimulation on the multifunctionality of T cells, by expression of the soluble extracellular part of PD-1 (sPD-1) or PD-L1 (sPD-L1) in human monocyte-derived DCs during antigen presentation. Expression, secretion and binding of these soluble molecules after mRNA electroporation were demonstrated. Modification of DCs with sPD-1 or sPD-L1 mRNA resulted in increased levels of the co-stimulatory molecule CD80 and a distinct cytokine profile, characterized by the secretion of IL-10 and TNF-α, respectively. Co-expression in DCs of sPD-1 and sPD-L1 with influenza virus nuclear protein 1 (Flu NP1) stimulated Flu NP1 memory T cells, with a significantly higher number of multifunctional T cells and increased cytokine secretion, while it did not induce regulatory T cells. These data provide a rationale for the inclusion of interfering sPD-1 or sPD-L1 in DC-based immunotherapeutic strategies.


Assuntos
Apresentação de Antígeno , Antígenos B7/imunologia , Células Dendríticas/imunologia , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T/imunologia , Antígenos B7/genética , Antígenos B7/metabolismo , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Células Dendríticas/metabolismo , Células HEK293 , Humanos , Interleucina-10/genética , Interleucina-10/metabolismo , Proteínas do Nucleocapsídeo , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
5.
Mol Hum Reprod ; 20(2): 168-77, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24217388

RESUMO

Gain of 20q11.21 is a chromosomal abnormality that is recurrently found in human pluripotent stem cells and cancers, strongly suggesting that this mutation confers a proliferative or survival advantage to these cells. In this work we studied three human embryonic stem cell (hESC) lines that acquired a gain of 20q11.21 during in vitro culture. The study of the mRNA gene expression levels of the loci located in the common region of duplication showed that HM13, ID1, BCL2L1, KIF3B and the immature form of the micro-RNA miR-1825 were up-regulated in mutant cells. ID1 and BCL2L1 were further studied as potential drivers of the phenotype of hESC with a 20q11.21 gain. We found no increase in the protein levels of ID1, nor the downstream effects expected from over-expression of this gene. On the other hand, hESC with a gain of 20q11.21 had on average a 3-fold increase of Bcl-xL (the anti-apoptotic isoform of BCL2L1) protein levels. The mutant hESC underwent 2- to 3-fold less apoptosis upon loss of cell-to-cell contact and were ∼2-fold more efficient in forming colonies from a single cell. The key role of BCL2L1 in this mutation was further confirmed by transgenic over-expression of BCL2L1 in the wild-type cells, leading to apoptosis-resistant cells, and BCL2L1-knock-down in the mutant hESC, resulting in a restoration of the wild-type phenotype. This resistance to apoptosis supposes a significant advantage for the mutant cells, explaining the high frequency of gains of 20q11.21 in human pluripotent stem cells.


Assuntos
Duplicação Cromossômica , Cromossomos Humanos Par 20 , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteína bcl-X/genética , Apoptose , Linhagem Celular , Sobrevivência Celular , Mapeamento Cromossômico , Células-Tronco Embrionárias/patologia , Expressão Gênica , Técnicas de Inativação de Genes , Loci Gênicos , Humanos , Células-Tronco Pluripotentes/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína bcl-X/antagonistas & inibidores , Proteína bcl-X/metabolismo
6.
Gene Ther ; 20(11): 1085-92, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23804077

RESUMO

Persistent activation of the transcription factor, signal transducer and activator of transcription 3 (Stat3) has been shown to mediate several oncogenic features in many types of cancers, including melanoma. In this study, we investigated whether lentiviral (LV) delivery of Stat3-targeting short hairpin RNA (shRNA; LV-shStat3) to K1735-C4 melanoma cells modulates antitumor immunity. Three shStat3 sequences, starting at the position 446, 830 and 1412, were cloned into a mir30 cassette. A shRNA with scrambled sequence served as a control. Transduction with LV-shStat3 resulted in downregulation of Stat3 in vitro. The latter coincided with low cell viability, a reduced expression of survivin and matrix metalloproteinase (MMP)-2. A single injection of LV-shStat3 in K1735-C4 tumors efficiently downregulated Stat3 in vivo and resulted in reduction of both vascular endothelial growth factor secretion and in myeloid-derived suppressor cell (MDSC) numbers. In contrast, we observed an increase in interleukin-6 and interferon-γ secretion, mature dendritic cells (DCs) and CD8(+) T cells. Both DCs and CD8(+) T cells displayed enhanced activity, whereas granulocytic MDSCs lost their suppressive capacity upon Stat3 downregulation. Importantly, a single injection of LV-shStat3 was sufficient to reduce tumor growth, hence prolong survival of tumor-bearing mice. These data demonstrate that Stat3 downregulation in melanoma reinvigorates existing antitumor immunity.


Assuntos
Melanoma Experimental/genética , Melanoma/genética , Fator de Transcrição STAT3/genética , Microambiente Tumoral/imunologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos , Células HEK293 , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Lentivirus/genética , Metaloproteinase 2 da Matriz/metabolismo , Melanoma/imunologia , Melanoma/metabolismo , Melanoma/terapia , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/administração & dosagem , Proteínas Repressoras/metabolismo , Fator de Transcrição STAT3/metabolismo , Survivina , Transdução Genética , Células Tumorais Cultivadas
7.
Curr Mol Med ; 13(4): 602-25, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22973872

RESUMO

Over the years, there has been an exponential increase in the number of gene therapy approaches that are under investigation for the treatment of cancer. This can be attributed to our growing understanding of the molecular mechanisms that contribute to the onset and maintenance of cancer as well as to the development of gene delivery vectors. In this review, we will focus on the use of lentiviral vectors (LVs) in immuno gene therapy of cancer, as these efficacious gene delivery vehicles have come to the fore front because of their many attractive features. LVs have been successfully applied to generate potent dendritic cell based anti-cancer vaccines and to deliver cancer-specific receptors to T-cells. Moreover, LVs are under investigation for the modulation of cancer cells. We will describe various strategies of this 'genuine' cancer gene therapy, amongst which transfer of suicide genes, modulation of pro- and anti-apoptotic molecules, strategies to optimize chemo- and radiotherapy, expression of molecules that affect angiogenesis or affect the immunogenicity of tumor cells. These will be discussed in view of our current knowledge of tumor immunology. Finally we will discuss some important issues and future directions to push the field forward.


Assuntos
Vetores Genéticos , Lentivirus/genética , Neoplasias/terapia , Terapia Genética , Humanos , Neoplasias/imunologia
8.
Gene Ther ; 19(12): 1133-40, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22241177

RESUMO

Lentiviral vectors (LVs) provide unique opportunities for the development of immunotherapeutic strategies, as they transduce a variety of cells in situ, including antigen-presenting cells (APCs). Engineering LVs to specifically transduce APCs is required to promote their translation towards the clinic. We report on the Nanobody (Nb) display technology to target LVs to dendritic cells (DCs) and macrophages. This innovative approach exploits the budding mechanism of LVs to incorporate an APC-specific Nb and a binding-defective, fusion-competent form of VSV.G in the viral envelope. In addition to production of high titer LVs, we demonstrated selective, Nb-dependent transduction of mouse DCs and macrophages both in vitro and in situ. Moreover, this strategy was translated to a human model in which selective transduction of in vitro generated or lymph node (LN)-derived DCs and macrophages, was demonstrated. In conclusion, the Nb display technology is an attractive approach to generate LVs targeted to specific cell types.


Assuntos
Células Apresentadoras de Antígenos , Marcação de Genes/métodos , Vetores Genéticos , Lentivirus/genética , Macrófagos , Anticorpos de Domínio Único , Animais , Técnicas de Transferência de Genes , Humanos , Camundongos , Transdução Genética
9.
Gene Ther ; 14(11): 847-62, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17361214

RESUMO

Lentiviral vectors have emerged as promising tools for both gene therapy and immunotherapy purposes. They exhibit several advantages over other viral systems in that they are less immunogenic and are capable of transducing a wide range of different cell types, including dendritic cells (DC). DC transduced ex vivo with a whole range of different (tumor) antigens were capable of inducing strong antigen-specific T-cell responses, both in vitro and in vivo. Recently, the administration of lentiviral vectors in vivo has gained substantial interest as an alternative method for antigen-specific immunization. This method offers a number of advantages over DC vaccines as the same lentivirus can in principle be used for all patients resulting in a significantly reduced cost and requirement for considerably less expertise for the generation and administration of lentiviral vaccines. By selectively targeting lentiviral vectors to, or restricting transgene expression in certain cell types, selectivity, safety and efficacy can be further improved. This review will focus on the use of direct administration of lentiviral vectors encoding tumor-associated antigens (TAA) for the induction of tumor-specific immune responses in vivo, with a special focus on problems related to the generation of large amounts of highly purified virus and specific targeting of antigen-presenting cells (APC).


Assuntos
Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Imunoterapia/métodos , Lentivirus/genética , Neoplasias/terapia , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Engenharia Genética , Vetores Genéticos/genética , Humanos , Modelos Animais , Neoplasias/imunologia
10.
Gene Ther ; 13(13): 1027-36, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16511519

RESUMO

The maturation state of dendritic cells (DCs) is an important determinant for the initiation and regulation of adaptive immune responses. In this study, we wanted to assess whether functional activation of human monocyte-derived DCs can be achieved by electroporation of an activation signal in the form of double-stranded (ds) RNA and whether simultaneous electroporation of the dsRNA with tumor antigen encoding mRNA can lead to the induction of a cytotoxic T-lymphocyte (CTL) response. Electroporation of immature DCs with poly(I:C(12)U), a dsRNA analogue, resulted in phenotypic as well as functional changes, indicative of DC maturation. Co-electroporation of DCs with both poly(I:C(12)U) and Melan-A/MART-1 encoding mRNA induced strong anti-Melan-A/MART-1 CD8(+) T-cell responses in vitro. Higher numbers of Melan-A/MART-1-specific CTLs were consistently obtained with poly(I:C(12)U)-activated DCs compared to DCs matured in the presence of an inflammatory cytokine cocktail. These results indicate that DC co-electroporation with both dsRNA and tumor antigen encoding mRNA induces fully activated and antigen-loaded DCs that promote antigen-specific CTL responses and may provide the basis for future immunotherapeutic strategies.


Assuntos
Antígenos de Neoplasias/genética , Linfócitos T CD8-Positivos/imunologia , Terapia Genética/métodos , Imunoterapia/métodos , Melanoma Experimental/terapia , Neoplasias Cutâneas/terapia , Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Testes Imunológicos de Citotoxicidade , Células Dendríticas/imunologia , Eletroporação/métodos , Citometria de Fluxo , Engenharia Genética , Humanos , Interferon gama/imunologia , Ativação Linfocitária , Antígeno MART-1 , Melanoma Experimental/imunologia , Proteínas de Neoplasias/genética , RNA/administração & dosagem , RNA de Cadeia Dupla/administração & dosagem , Neoplasias Cutâneas/imunologia , Células Tumorais Cultivadas
11.
Gene Ther ; 13(7): 630-40, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16355115

RESUMO

Ex vivo lentivirally transduced dendritic cells (DC) have been described to induce CD8+ and CD4+ T-cell responses against various tumor-associated antigens (TAAs) in vitro and in vivo. We report here that direct administration of ovalbumin (OVA) encoding lentiviral vectors caused in vivo transduction of cells that were found in draining lymph nodes (LNs) and induced potent anti-OVA cytotoxic T cells similar to those elicited by ex vivo transduced DC. The cytotoxic T-lymphocyte (CTL) response following direct injection of lentiviral vectors was highly effective in eliminating target cells in vivo up to 30 days after immunization and was efficiently recalled after a boost immunization. Injection of lentiviral vectors furthermore activated OVA-specific CD4+ T cells and this CD4 help was shown to be necessary for an adequate primary and memory CTL response. When tested in therapeutic tumor experiments with OVA+ melanoma cells, direct administration of lentiviral vectors slowed down tumor growth to a comparable extent with the highest dose of ex vivo transduced DC. Taken together, these data indicate that direct in vivo administration of lentiviral vectors encoding TAAs has strong potential for anticancer vaccination.


Assuntos
Células Dendríticas/imunologia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Imunoterapia/métodos , Lentivirus/genética , Neoplasias/terapia , Animais , Antígenos Glicosídicos Associados a Tumores/imunologia , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular Tumoral , Células Dendríticas/virologia , Feminino , Memória Imunológica , Interferon gama/imunologia , Linfonodos/imunologia , Linfonodos/virologia , Ativação Linfocitária , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Ovalbumina/genética , Ovalbumina/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Citotóxicos/imunologia , Transdução Genética/métodos
12.
Gene Ther ; 12(9): 772-82, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15750615

RESUMO

Until now, studies utilizing mRNA electroporation as a tool for the delivery of tumor antigens to human monocyte-derived dendritic cells (DC) have focused on DC electroporated in an immature state. Immature DC are considered to be specialized in antigen capture and processing, whereas mature DC present antigen and have an increased T-cell stimulatory capacity. Therefore, the consensus has been to electroporate DC before maturation. We show that the transfection efficiency of DC electroporated either before or after maturation was similarly high. Both immature and mature electroporated DC, matured in the presence of an inflammatory cytokine cocktail, expressed mature DC surface markers and preserved their capacity to secrete cytokines and chemokines upon CD40 ligation. In addition, both immature and mature DC can be efficiently cryopreserved before or after electroporation without deleterious effects on viability, phenotype or T-cell stimulatory capacity including in vitro antigen-specific T-cell activation. However, DC electroporated after maturation are more efficient in in vitro migration assays and at least as effective in antigen presentation as DC electroporated before maturation. These results are important for vaccination strategies where an optimal antigen presentation by DC after migration to the lymphoid organs is crucial.


Assuntos
Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Eletroporação , Apresentação de Antígeno , Diferenciação Celular/imunologia , Sobrevivência Celular , Criopreservação , Citocinas/imunologia , Humanos , Imunofenotipagem , Ativação Linfocitária/imunologia , Teste de Cultura Mista de Linfócitos , RNA Mensageiro/metabolismo , Linfócitos T/imunologia , Transfecção
13.
Diabetologia ; 47(6): 998-1008, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15184981

RESUMO

AIMS/HYPOTHESIS: In the human pancreas, a close topographic relationship exists between duct cells and beta cells. This explains the high proportion of duct cells in isolated human islet preparations. We investigated whether human duct cells are a source of TNFalpha-mediated interactions with beta cells and immune cells. This cytokine has been implicated in the development of autoimmune diabetes in mice. METHODS: Human duct cells were isolated from donor pancreases and examined for their ability to produce TNFalpha following a stress-signalling pathway. Duct-cell-released TNFalpha was tested for its in vitro effects on survival of human beta cells and on activation of human dendritic cells. RESULTS: Exposure of human pancreatic duct cells to interleukin-1beta (IL-1beta) induces TNFalpha gene expression, synthesis of the 26,000 M(r) TNFalpha precursor and conversion to the 17,000 M(r) mature form, which is rapidly released. This effect is NO-independent and involves p38 MAPK and NF-kappaB signalling. Duct-cell-released TNFalpha contributed to cytokine-induced apoptosis of isolated human beta cells. It also induced activation of human dendritic cells. CONCLUSIONS/INTERPRETATION: Human pancreatic duct cells are a potential source of TNFalpha that can cause apoptosis of neighbouring beta cells and initiate an immune response through activation of dendritic cells. They may thus actively participate in inflammatory and immune processes that threaten beta cells during development of diabetes or after human islet cell grafts have been implanted.


Assuntos
Células Dendríticas/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ductos Pancreáticos/metabolismo , Fator de Necrose Tumoral alfa/efeitos adversos , Fator de Necrose Tumoral alfa/metabolismo , Apoptose , Bélgica , Brefeldina A/farmacologia , Células Cultivadas , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Células Dendríticas/efeitos dos fármacos , Humanos , Imuno-Histoquímica/métodos , Interleucina-1/química , Interleucina-1/metabolismo , Interleucina-1/farmacologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/fisiopatologia , Queratinas/química , Queratinas/imunologia , NF-kappa B/metabolismo , Ductos Pancreáticos/patologia , Ductos Pancreáticos/ultraestrutura , Precursores de Proteínas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...