Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 17(1): 140-149, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29079709

RESUMEN

Targeted therapy against VEGF and mTOR pathways has been established as the standard-of-care for metastatic clear cell renal cell carcinoma (ccRCC); however, these treatments frequently fail and most patients become refractory requiring subsequent alternative therapeutic options. Therefore, development of innovative and effective treatments is imperative. About 80%-90% of ccRCC tumors express an inactive mutant form of the von Hippel-Lindau protein (pVHL), an E3 ubiquitin ligase that promotes target protein degradation. Strong genetic and experimental evidence supports the correlate that pVHL functional loss leads to the accumulation of the transcription factor hypoxia-inducible factor 2α (HIF2α) and that an overabundance of HIF2α functions as a tumorigenic driver of ccRCC. In this report, we describe an RNAi therapeutic for HIF2α that utilizes a targeting ligand that selectively binds to integrins αvß3 and αvß5 frequently overexpressed in ccRCC. We demonstrate that functional delivery of a HIF2α-specific RNAi trigger resulted in HIF2α gene silencing and subsequent tumor growth inhibition and degeneration in an established orthotopic ccRCC xenograft model. Mol Cancer Ther; 17(1); 140-9. ©2017 AACR.


Asunto(s)
Carcinoma de Células Renales/terapia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , ARN Interferente Pequeño/administración & dosificación , Animales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Femenino , Silenciador del Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Integrina alfaVbeta3/metabolismo , Ratones , Ratones Desnudos , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Vitronectina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mol Ther Nucleic Acids ; 1: e46, 2012 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-23344238

RESUMEN

Bispecific antibodies (bsAbs) that bind to cell surface antigens and to digoxigenin (Dig) were used for targeted small interfering RNA (siRNA) delivery. They are derivatives of immunoglobulins G (IgGs) that bind tumor antigens, such as Her2, IGF1-R, CD22, and LeY, with stabilized Dig-binding variable domains fused to the C-terminal ends of the heavy chains. siRNA that was digoxigeninylated at its 3'end was bound in a 2:1 ratio to the bsAbs. These bsAb-siRNA complexes delivered siRNAs specifically to cells that express the corresponding antigen as demonstrated by flow cytometry and confocal microscopy. The complexes internalized into endosomes and Dig-siRNAs separated from bsAbs, but Dig-siRNA was not released into the cytoplasm; bsAb-targeting alone was thus not sufficient for effective mRNA knockdown. This limitation was overcome by formulating the Dig-siRNA into nanoparticles consisting of dynamic polyconjugates (DPCs) or into lipid-based nanoparticles (LNPs). The resulting complexes enabled bsAb-targeted siRNA-specific messenger RNA (mRNA) knockdown with IC(50) siRNA values in the low nanomolar range for a variety of bsAbs, siRNAs, and target cells. Furthermore, pilot studies in mice bearing tumor xenografts indicated mRNA knockdown in endothelial cells following systemic co-administration of bsAbs and siRNA formulated in LNPs that were targeted to the tumor vasculature.Molecular Therapy - Nucleic Acids (2012) 1, e45; doi:10.1038/mtna.2012.39; published online 18 September 2012.

3.
J Leukoc Biol ; 89(4): 625-38, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21248148

RESUMEN

The hu14.18-IL2 (EMD 273063) IC, consisting of a GD(2)-specific mAb genetically engineered to two molecules of IL-2, is in clinical trials for treatment of GD(2)-expressing tumors. Anti-tumor activity of IC in vivo and in vitro involves NK cells. We studied the kinetics of retention of IC on the surface of human CD25(+)CD16(-) NK cell lines (NKL and RL12) and GD(2)(+) M21 melanoma after IC binding to the cells via IL-2R and GD(2), respectively. For NK cells, ∼ 50% of IC was internalized by 3 h and ∼ 90% by 24 h of cell culture. The decrease of surface IC levels on NK cells correlated with the loss of their ability to bind to tumor cells and mediate antibody-dependent cellular cytotoxicity in vitro. Unlike NK cells, M21 cells retained ∼ 70% of IC on the surface following 24 h of culture and maintained the ability to become conjugated and lysed by NK cells. When NKL cells were injected into M21-bearing SCID mice, IT delivery of IC augmented NK cell migration into the tumor. These studies demonstrate that once IC binds to the tumor, it is present on the tumor surface for a prolonged time, inducing the recruitment of NK cells to the tumor site, followed by tumor cell killing.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos , Interleucina-2/farmacología , Células Asesinas Naturales/inmunología , Melanoma/inmunología , Melanoma/terapia , Animales , Citometría de Flujo , Humanos , Inmunofenotipificación , Células Asesinas Naturales/efectos de los fármacos , Melanoma/genética , Ratones , Ratones SCID , Células Tumorales Cultivadas
4.
Chem Biol ; 14(9): 1065-77, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17884638

RESUMEN

We have investigated a rapidly reversible hydrophobization of therapeutic agents for improving first-pass uptake in locoregional drug therapy. This approach involves the attachment of a hydrophobic moiety to the drug by highly labile chemical linkages that rapidly hydrolyze upon injection. Hydrophobization drastically enhances cell-membrane association of the prodrug and, consequently, drug uptake, while the rapid lability protects nontargeted tissues from exposure to the highly active agent. Using the membrane-impermeable DNA intercalator propidium iodide, and melphalan, we report results from in vitro cellular internalization and toxicity studies. Additionally, we report in vivo results after a single liver arterial bolus injection, demonstrating both tumor targeting and increased survival in a mouse tumor model.


Asunto(s)
Antineoplásicos/administración & dosificación , Interacciones Hidrofóbicas e Hidrofílicas , Profármacos/química , Profármacos/farmacocinética , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Melfalán/administración & dosificación , Propidio , Resultado del Tratamiento
5.
Cancer Immunol Immunother ; 56(11): 1765-74, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17426968

RESUMEN

Therapeutic treatment with hu14.18-IL-2 immunocytokine (IC) or Flt3-L (FL) protein is initially effective at resolving established intradermal NXS2 neuroblastoma tumors in mice. However, many treated animals develop recurrent disease. We previously found that tumors recurring following natural killer (NK) mediated IC treatment show augmented MHC class I expression, while the tumors that recurred following T cell dependent Flt3-L treatment exhibited decreased MHC class I expression. We hypothesized that this divergent MHC modulation on recurrent tumors was due to therapy-specific immunoediting. We further postulated that combining IC and Flt3-L treatments might decrease the likelihood of recurrent disease by preventing MHC modulation as a mechanism for immune escape. We now report that combinatorial treatment of FL plus hu14.18-IL-2 IC provides greater antitumor benefit than treatment with either alone, suppressing development of recurrent disease. We administered FL by gene therapy using a clinically relevant approach: hydrodynamic limb vein (HLV) delivery of DNA for transgene expression by myofibers. Delivery of FL DNA by HLV injection in mice resulted in systemic expression of >10 ng/ml of FL in blood at day 3, and promoted up to a fourfold and tenfold increase in splenic NK and dendritic cells (DCs), respectively. Furthermore, the combination of FL gene therapy plus suboptimal IC treatment induced a greater expansion in the absolute number of splenic NK and DCs than achieved by individual component treatments. Mice that received combined FL gene therapy plus IC exhibited complete and durable resolution of established NXS2 tumors, and demonstrated protection from subsequent rechallenge with NXS2 tumor.


Asunto(s)
Terapia Genética , Memoria Inmunológica , Interleucina-2/farmacología , Proteínas de la Membrana/farmacología , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/metabolismo , Adyuvantes Inmunológicos/farmacología , Animales , Antineoplásicos/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/metabolismo , Bazo/inmunología , Factores de Tiempo , Escape del Tumor/inmunología
6.
Mol Ther ; 15(2): 422-30, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17235322

RESUMEN

Tumor-associated antigens (TAA) are typically poorly immunogenic "self" antigens. An effective strategy to break tolerance and induce antitumor immunity is by genetic vaccination, employing the orthologous TAA-sequence from a different species. We recently developed a clinically relevant approach for intravascular hydrodynamic limb vein (HLV) delivery of nucleic acids to skeletal muscle. Using the human gp100 xenogeneic TAA in the murine B16 melanoma model, we show that genetic vaccination of mice by HLV plasmid DNA delivery was highly effective at breaking tolerance against the homologous murine gp100 (mgp100) TAA and induced prophylactic antitumor protection. HLV vaccination resulted in an anti-hgp100 humoral and cellular response, with 4-5% of CD8(+) T cells being gp100(25-33)-epitope-specific. Vaccinated animals demonstrated in vivo cytolytic activity against human and mgp100(25-33) peptide-pulsed targets. Antitumor immunity could be adoptively transferred by splenocytes from human gp100-vaccinated animals. Furthermore, a durable antitumor memory response was established as approximately 3% of CD8(+) T cells were gp100(25-33) antigen-specific in mice 6 months after vaccination. Following a single HLV human gp100 DNA boost, this level increased to approximately 17% and protected animals from subsequent B16 tumor rechallenge. Our results warrant further consideration of HLV as a clinically relevant method for cancer gene therapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Melanoma Experimental/inmunología , Vacunas de ADN/inmunología , Animales , Western Blotting , Linfocitos T CD8-positivos/inmunología , Células COS , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Chlorocebus aethiops , Epítopos de Linfocito T/inmunología , Femenino , Citometría de Flujo , Miembro Posterior/irrigación sanguínea , Humanos , Inyecciones Intravenosas/métodos , Melanoma Experimental/patología , Melanoma Experimental/prevención & control , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Vacunación/métodos , Vacunas de ADN/administración & dosificación , Antígeno gp100 del Melanoma
7.
Biotechniques ; 40(2): 199-208, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16526410

RESUMEN

Genetic immunization is an attractive approach to generate antibodies because native proteins are expressed in vivo with normal posttranscriptional modifications, avoiding time-consuming and costly antigen isolation or synthesis. Hydrodynamic tail or limb vein delivery of naked plasmid DNA expression vectors was used to induce antigen-specific antibodies in mice, rats, and rabbits. Both methods allowed the efficient generation of high-titer, antigen-specific antibodies with an overall success rate of Western detectable antibodies of 78% and 92%, respectively. High-titer antibodies were typically present after 3 hydrodynamic tail vein plasmid DNA deliveries, 5 weeks after the initial injection (i.e., prime). For hydrodynamic limb vein plasmid DNA delivery, two deliveries were sufficient to induce high-titer antibody levels. Tail vein delivery was less successful at generating antibodies directed against secreted proteins as compared with limb vein delivery. Material for screening was generated by,transfection of the immunization vector into mammalian cell lines. The cell line (COS-7) that produced the highest level of antigen expression performed best in Western blot analysis screens. In summary, intravenous delivery of antigen-expressing plasmid DNA vectors is an effective genetic immunization method for the induction of antigen-specific antibodies in small and large research animals.


Asunto(s)
Formación de Anticuerpos , Técnicas de Transferencia de Gen , Plásmidos , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Western Blotting , Antígenos CD4/genética , Células COS , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Vectores Genéticos , Células HeLa , Humanos , Hibridomas , Inmunohistoquímica , Inyecciones Intravenosas , Ratones , Ratones Endogámicos ICR , Conejos , Ratas , Ratas Sprague-Dawley , Transfección , Vacunas de ADN/genética
8.
Clin Cancer Res ; 10(14): 4839-47, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15269160

RESUMEN

Established s.c. NXS2 murine neuroblastoma tumors exhibited transient resolution after suboptimal therapy using the hu14.18-IL2 immunocytokine (IC). The hu14.18-IL2 IC is a fusion protein that has linked a molecule of interleukin 2 (IL-2) to the COOH terminus of each of the IgG heavy chains on the humanized anti-GD(2) monoclonal antibody hu14.18. To induce more potent and longer lasting in vivo antitumor effects, we tested hu14.18-IL2 IC in a regimen combining it with constant infusion IL-2 in NXS2 tumor-bearing mice. The addition of the constant infusion IL-2 augmented the antitumor response induced by treatment with the hu14.18-IL2 IC in animals with experimentally induced hepatic metastases and in animals bearing localized s.c. tumors. The combined treatment induced prolonged tumor eradication in most animals bearing s.c. tumors and involved both natural killer cells and T cells. The enhanced ability of this combined treatment to prevent tumor recurrence was not observed when a larger dose of hu14.18-IL2 IC, similar in IL-2 content to the IC plus systemic IL-2 regimen, was tested as single-agent therapy. Animals showing prolonged tumor eradication of established tumors after the combined hu14.18-IL2 plus IL-2 regimen exhibited a protective T-cell-dependent antitumor memory response against NXS2 rechallenge.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Interleucina-2/farmacología , Neuroblastoma/prevención & control , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de los fármacos , Femenino , Gangliósidos/inmunología , Humanos , Interleucina-2/química , Interleucina-2/uso terapéutico , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas Experimentales/prevención & control , Neoplasias Hepáticas Experimentales/secundario , Ratones , Ratones Endogámicos , Metástasis de la Neoplasia/prevención & control , Neuroblastoma/patología , Neuroblastoma/terapia , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Bazo/citología , Bazo/inmunología , Factores de Tiempo
9.
Cancer Immunol Immunother ; 53(1): 41-52, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14504825

RESUMEN

We evaluated recurrent NXS2 neuroblastoma tumors that developed following NK- or T-cell-mediated immunotherapy in tumor-bearing mice. Recurrent tumors developed following an NK-dependent antitumor response using a suboptimal dose of hu14.18-IL2, a humanized IL-2 immunocytokine targeted to the GD(2)-ganglioside. This treatment initially induced complete resolution of measurable tumor in the majority of mice, followed, however, by delayed tumor recurrence in some mice. These recurrent NXS2 tumors revealed markedly enhanced (> fivefold) MHC class I antigen expression when compared with NXS2 tumors growing in PBS-treated control mice. A similar level of enhanced MHC class I antigen-expression could be induced on NXS2 cells in vitro by culturing with interferon gamma, and was associated with reduced susceptibility to both NK-cell-mediated tumor cell lysis and antibody-dependent cellular cytotoxicity in vitro. In contrast, Flt3-ligand treatment of NXS2-bearing mice induced a protective T-cell-dependent antitumor memory response. Recurrent NXS2 tumors that developed following Flt3-L therapy revealed a decreased expression of MHC class I antigens. While NXS2 tumors are susceptible to in vivo destruction following either hu14.18-IL2 or Flt3-ligand immunotherapies, these results suggest that some tumor cells may be selected to survive and progress by expressing either higher or lower levels of MHC class I antigen in order to resist either NK- or T-cell-mediated antitumor responses, respectively.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoterapia , Recurrencia Local de Neoplasia/terapia , Neuroblastoma/terapia , Neoplasias Cutáneas/terapia , Animales , Técnicas de Cocultivo , Femenino , Humanos , Inyecciones Subcutáneas , Interleucina-2/uso terapéutico , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/uso terapéutico , Ratones , Recurrencia Local de Neoplasia/inmunología , Neuroblastoma/inmunología , Neoplasias Cutáneas/inmunología , Linfocitos T/inmunología , Células Tumorales Cultivadas , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
10.
Viral Immunol ; 15(1): 155-63, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11952137

RESUMEN

Encephalomyocarditis virus (EMCV) and Mengo virus are highly virulent murine cardioviruses that are found in abundant quantities in the spleen and lymph nodes after infection. T lymphocytes are pivotal mediators of humoral and cellular immunity against cardioviral challenge, and are highly suspect candidates of EMCV and Mengo virus infection. We found T lymphocyte-like cell lines CTLL-2, EL-4, LY1+2/9, and LBRM33 were susceptible to productive viral infection and exhibited cytopathology after infection with virulent EMCV-R or attenuated Mengo virus strains vMC0 and vMC24. Flow cytometric analysis demonstrated progressive intracellular accumulation of viral proteins, such as the replication-dependent 3D viral polymerase, in EL-4 cells during infection. Conversely, freshly isolated and mitogen-stimulated CD4+ and CD8+ T cells were resistant to productive infection with these viruses, exhibiting no viral-induced cytopathic effects or intracellular presence of viral proteins. These data indicate that although T-lymphocyte-like tumor cell lines are highly susceptible to viral infection and cytopathic effects, primary/freshly isolated T cells are resistant to infection by EMCV-R or Mengo virus.


Asunto(s)
Virus de la Encefalomiocarditis/fisiología , Mengovirus/fisiología , Linfocitos T/virología , Animales , Línea Celular , Efecto Citopatogénico Viral , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Proteínas Virales/análisis , Replicación Viral , eIF-2 Quinasa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA