Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(34): 13791-6, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23918357

RESUMO

Antibody cancer therapies rely on systemically accessible targets and suitable antibodies that exert a functional activity or deliver a payload to the tumor site. Here, we present proof-of-principle of in vivo selection of human antibodies in tumor-bearing mice that identified a tumor-specific antibody able to deliver a payload and unveils the target antigen. By using an ex vivo enrichment process against freshly disaggregated tumors to purge the repertoire, in combination with in vivo biopanning at optimized phage circulation time, we have identified a human domain antibody capable of mediating selective localization of phage to human prostate cancer xenografts. Affinity chromatography followed by mass spectrometry analysis showed that the antibody recognizes the proteasome activator complex PA28. The specificity of soluble antibody was confirmed by demonstrating its binding to the active human PA28αß complex. Whereas systemically administered control phage was confined in the lumen of blood vessels of both normal tissues and tumors, the selected phage spread from tumor vessels into the perivascular tumor parenchyma. In these areas, the selected phage partially colocalized with PA28 complex. Furthermore, we found that the expression of the α subunit of PA28 [proteasome activator complex subunit 1 (PSME1)] is elevated in primary and metastatic human prostate cancer and used anti-PSME1 antibodies to show that PSME1 is an accessible marker in mouse xenograft tumors. These results support the use of PA28 as a tumor marker and a potential target for therapeutic intervention in prostate cancer.


Assuntos
Anticorpos Antineoplásicos/imunologia , Biomarcadores Tumorais/imunologia , Imunoterapia/métodos , Proteínas Musculares/metabolismo , Neoplasias da Próstata/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Anticorpos Antineoplásicos/metabolismo , Especificidade de Anticorpos , Western Blotting , Técnicas de Visualização da Superfície Celular , Cromatografia de Afinidade , Cromatografia Líquida , Sistemas de Liberação de Medicamentos/métodos , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Humanos , Imuno-Histoquímica , Imunoprecipitação , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/terapia , Estatísticas não Paramétricas , Espectrometria de Massas em Tandem
2.
J Biol Chem ; 289(4): 2457-68, 2014 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-24307174

RESUMO

Pericytes and mesenchymal stem cells (MSCs) are ontogenically related, and in fact, no significant phenotypic differences could be observed by flow cytometry. Transcriptome analysis of human pericytes and MSCs revealed that 43 genes were up-regulated more than 10-fold in pericytes compared with MSCs. Identification of Toll-like receptor 4 (TLR4) as one of the most abundant RNA species in pericytes with respect to MSCs and confirmation of TLR4 expression on the cell surface led us to obtain a comprehensive overview of the expression program of lipopolysaccharide (LPS)-stimulated pericytes. Transcriptional profiling of LPS-treated cells revealed that 22 genes were up-regulated more than 5-fold. Of them, 10 genes encoded chemokines and cytokines (CXCL10, CCL20, IL8, CXCL1, IL6, CCL2, IL1B, CXCL2, IL1A, and CXCL6), and three genes encoded adhesion molecules (ICAM1, VCAM1, and SELE). LPS induced nuclear translocation of the transcription factor NF-κB in stimulated pericytes. Moreover, inhibition of NF-κB activation by SC-514 blocked LPS-induced up-regulation of a subset of chemokine genes, confirming the key role of NF-κB in LPS signaling in pericytes. At the protein level, we assessed the secretion of the proinflammatory cytokines and chemokines IL-6, IL-8, CXCL1, CXCL2, CXCL3, and CCL2 not only after LPS treatment but also in HMGB1-stimulated pericytes. Up-regulation of the adhesion molecules ICAM-1 and VCAM-1 resulted in an increased adhesion of peripheral blood leukocytes to an LPS-treated pericyte monolayer. The role of pericytes in the inflammatory context has been scarcely addressed; according to these results, pericytes should be considered as active players in the inflammatory cascade with potential physiopathological implications.


Assuntos
Moléculas de Adesão Celular/biossíntese , Citocinas/biossíntese , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Pericitos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/agonistas , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/imunologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Moléculas de Adesão Celular/imunologia , Núcleo Celular/imunologia , Núcleo Celular/metabolismo , Células Cultivadas , Técnicas de Cocultura , Citocinas/imunologia , Humanos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Leucócitos/citologia , Leucócitos/imunologia , Leucócitos/metabolismo , NF-kappa B/imunologia , Pericitos/citologia , Pericitos/imunologia , Transdução de Sinais/imunologia , Tiofenos/farmacologia , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
4.
Curr Gene Ther ; 13(4): 282-90, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23773178

RESUMO

Monoclonal antibodies are successfully used in the clinic. However, repeated high-dose bolus injections imply high costs. As an alternative to recombinant protein administration, gene therapy may provide a novel mean for systemic delivery of monoclonal antibodies. This strategy has been used in preclinical studies of a wide variety of pathological conditions, including cancer, infectious diseases, drug addiction, retinal neovascularisation and Alzheimer's disease. The two main gene therapy approaches are based on direct gene delivery (using viral or non-viral vectors) or on inoculation of ex vivo genetically modified cells (autologous or allogenic). Viral vectors are highly efficient as gene delivery vehicles and have been tested in numerous clinical trials, but still raise concerns about safety and limitation of the therapeutic effect due to immune responses against viral antigens. On the other hand, use of standard non-viral vectors has been limited by their low transduction efficiency. Practical application of ex vivo genetically modified cells would imply the availability of stocks of "ready-to-use" gene-modified allogenic cells that should be protected from the host immune system. Actually, this field did not meet the expectation raised initially, mainly because of difficulties with obtaining sustained therapeutic plasma levels in animal models. However, this situation is changing rapidly and the therapeutic potential of these approaches is getting closer to clinical applications. This review focuses on the current achievements in preclinical studies, as well as the challenges and future prospects of antibody gene therapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Doenças Transmissíveis/terapia , Neoplasias/terapia , Proteínas Recombinantes/uso terapêutico , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Doenças Transmissíveis/imunologia , Doenças Transmissíveis/patologia , Terapia Genética , Humanos , Neoplasias/imunologia , Neoplasias/patologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
5.
Med Oncol ; 30(2): 572, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23579861

RESUMO

The anti-VEGF monoclonal antibody bevacizumab was approved in 2004 as a first-line treatment for metastatic colorectal cancer (CRC) in combination with chemotherapy and provided proof of principle for antiangiogenic therapy. However, there is no biomarker that can help to select patients who may benefit from bevacizumab in order to improve cost-effectiveness and therapeutic outcomes. The aim of this study was to compare gene expression profiles in CRC patients treated with bevacizumab who responded to the treatment with those that did not respond, in an effort to identify potential predictive biomarkers. RNA isolated from formalin-fixed paraffin-embedded tumor specimens of patients treated with bevacizumab was subjected to gene expression analysis with quantitative RT-PCR arrays profiling 84 genes implicated in the angiogenic process. Data were validated at the protein level using immunohistochemistry. We identified a gene, EPHB4, whose expression was significantly increased in nonresponders (p = 0.048, Mann-Whitney test). Furthermore, high EPHB4 tumor levels were associated with decreased median overall survival (16 months vs 48, Log-rank p = 0.012). This was not observed in a control group of CRC patients treated only with chemotherapy, suggesting that EPHB4 constitutes a potential predictive biomarker and not a mere prognostic one. These data support the notion of a potential synergy between EPHB4-EFNB2 and VEGF-VEGFR pathways, making patients with high EPHB4 expression more resistant to VEGF blocking. Therefore, determination of EPHB4 levels in CRC samples could be useful for the prediction of response to bevacizumab.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Biomarcadores Tumorais/biossíntese , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Receptor EphB4/genética , Adulto , Idoso , Bevacizumab , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Prognóstico , Receptor EphB4/biossíntese , Receptor EphB4/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
PLoS One ; 8(8): e72957, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951338

RESUMO

Metastatic breast cancer is the leading cause of death by malignancy in women worldwide. Tumor metastasis is a multistep process encompassing local invasion of cancer cells at primary tumor site, intravasation into the blood vessel, survival in systemic circulation, and extravasation across the endothelium to metastasize at a secondary site. However, only a small percentage of circulating cancer cells initiate metastatic colonies. This fact, together with the inaccessibility and structural complexity of target tissues has hampered the study of the later steps in cancer metastasis. In addition, most data are derived from in vivo models where critical steps such as intravasation/extravasation of human cancer cells are mediated by murine endothelial cells. Here, we developed a new mouse model to study the molecular and cellular mechanisms underlying late steps of the metastatic cascade. We have shown that a network of functional human blood vessels can be formed by co-implantation of human endothelial cells and mesenchymal cells, embedded within a reconstituted basement membrane-like matrix and inoculated subcutaneously into immunodeficient mice. The ability of circulating cancer cells to colonize these human vascularized organoids was next assessed in an orthotopic model of human breast cancer by bioluminescent imaging, molecular techniques and immunohistological analysis. We demonstrate that disseminated human breast cancer cells efficiently colonize organoids containing a functional microvessel network composed of human endothelial cells, connected to the mouse circulatory system. Human breast cancer cells could be clearly detected at different stages of the metastatic process: initial arrest in the human microvasculature, extravasation, and growth into avascular micrometastases. This new mouse model may help us to map the extravasation process with unprecedented detail, opening the way for the identification of relevant targets for therapeutic intervention.


Assuntos
Neoplasias da Mama/patologia , Metástase Neoplásica/patologia , Células Neoplásicas Circulantes/patologia , Organoides/irrigação sanguínea , Organoides/patologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Nus
7.
PLoS One ; 7(6): e39097, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22723936

RESUMO

Laminins are large heterotrimeric cross-shaped extracellular matrix glycoproteins with terminal globular domains and a coiled-coil region through which the three chains are assembled and covalently linked. Laminins are key components of basement membranes, and they serve as attachment sites for cell adhesion, migration and proliferation. In this work, we produced a recombinant fragment comprising the entire laminin coiled-coil of the α1-, ß1-, and γ1-chains that assemble into a stable heterotrimeric coiled-coil structure independently of the rest of the molecule. This domain was biologically active and not only failed to serve as a substrate for cell attachment, spreading and focal adhesion formation but also inhibited cell adhesion to laminin when added to cells in a soluble form at the time of seeding. Furthermore, gene array expression profiling in cells cultured in the presence of the laminin coiled-coil domain revealed up-regulation of genes involved in cell motility and invasion. These findings were confirmed by real-time quantitative PCR and zymography assays. In conclusion, this study shows for the first time that the laminin coiled-coil domain displays anti-adhesive functions and has potential implications for cell migration during matrix remodeling.


Assuntos
Movimento Celular/genética , Laminina/genética , Laminina/metabolismo , Domínios e Motivos de Interação entre Proteínas/fisiologia , Multimerização Proteica , Animais , Adesão Celular/genética , Linhagem Celular Tumoral , Ativação Enzimática , Expressão Gênica , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Laminina/química , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA