Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Crohns Colitis ; 9(8): 647-58, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25987350

RESUMEN

BACKGROUND AND AIMS: We investigated oral delivery of transforming growth factor beta 1 [TGFß]- and all-trans retinoic acid [ATRA]-loaded microspheres as therapy for gut inflammation in murine models of inflammatory bowel disease [IBD]. METHODS: ATRA and TGFß were separately encapsulated in poly [lactic-co-glycolic] acid or polylactic acid microspheres [respectively]. TGFß was encapsulated using proprietary phase-inversion nanoencapsulation [PIN] technology. RESULTS: PIN particles provided sustained release of bioactive protein for at least 4 days and were stable for up to 52 weeks when stored at either 4(0)C or -20(0)C. In the SCID mouse CD4 + CD25- T cell transfer model of IBD, oral treatment starting at disease onset prevented weight loss, significantly reduced average disease score [~ 50%], serum amyloid A levels [~ 5-fold], colon weight-to-length ratio [~ 50%], and histological score [~ 5-fold]. CONCLUSIONS: Both agents given together outperformed either separately. Highest TGFß doses and most frequent dose schedule were most effective. Activity was associated with a significant increase [45%] in Foxp3 expression by colonic lamina propria CD4+ CD25+ T-cells. Activity was also demonstrated in dextran sulphate sodium-induced colitis. The data support development of the combination product as a novel, targeted immune based therapy for treatment for IBD.


Asunto(s)
Antiinflamatorios/administración & dosificación , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Factor de Crecimiento Transformador beta/administración & dosificación , Tretinoina/administración & dosificación , Administración Oral , Animales , Antiinflamatorios/uso terapéutico , Biomarcadores/metabolismo , Esquema de Medicación , Quimioterapia Combinada , Femenino , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Microesferas , Factor de Crecimiento Transformador beta/uso terapéutico , Resultado del Tratamiento , Tretinoina/uso terapéutico
2.
Cancer Res ; 74(19): 5377-85, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25228656

RESUMEN

Immune dysregulation drives the pathogenesis of chronic inflammatory, autoimmune, and dysplastic disorders. While often intended to address localized pathology, most immune modulatory therapies are administered systemically and carry inherent risk of multiorgan toxicities. Here, we demonstrate, in a murine model of spontaneous gastrointestinal polyposis, that site-specific uptake of orally administered IL10 microparticles ameliorates local and systemic disease to enhance survival. Mechanistic investigations showed that the therapeutic benefit of this treatment derived from neutralization of disease-promoting FoxP3(+)RoRγt(+)IL17(+) pathogenic T-regulatory cells (pgTreg), with a concomitant restoration of FoxP3(+)RoRγt(-)IL17(-) conventional T-regulatory cells (Treg). These findings provide a proof-of-principle for the ability of an oral biologic to restore immune homeostasis at the intestinal surface. Furthermore, they implicate local manipulation of IL10 as a tractable therapeutic strategy to address the inflammatory sequelae associated with mucosal premalignancy.


Asunto(s)
Poliposis Adenomatosa del Colon/prevención & control , Interleucina-10/farmacología , Pruebas de Neutralización , Linfocitos T Reguladores/efectos de los fármacos , Administración Oral , Secuencia de Bases , Cartilla de ADN , Humanos , Interleucina-10/administración & dosificación , Depleción Linfocítica , Reacción en Cadena de la Polimerasa , Linfocitos T Reguladores/inmunología
3.
J Assoc Res Otolaryngol ; 13(3): 423-35, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22310933

RESUMEN

A majority of T cells from chronic inflammatory tissues derived from patients with nasal polyposis were found to express an effector memory phenotype. We report here that these memory T cells failed to activate NF-κB in response to TCR stimulation but responded normally when the proximal TCR signaling molecules were bypassed with PMA and ionomycin. The dysfunction of these cells was associated with a decrease in the phosphorylation of several TCR proximal signaling molecules including ZAP70, Lck and SLP-76. In addition to the disruption in the TCR signaling pathway, the nasal polyp-associated T cells were shown to have a defect in their ability to translocate LAMP-1 to the cell surface. The results presented here establish that the phenotype and anergy of the T cells in the nasal polyp are similar to those which is seen in memory T cells derived from human tumors and other sites of chronic inflammation.


Asunto(s)
Antígenos CD28/metabolismo , Complejo CD3/metabolismo , Pólipos Nasales/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/fisiología , Ionóforos de Calcio , Estudios de Casos y Controles , Citometría de Flujo , Humanos , Memoria Inmunológica , Inmunofenotipificación , Ionomicina , Activación de Linfocitos , Proteínas de Membrana de los Lisosomas/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Fenotipo , Fosforilación , Transducción de Señal , Linfocitos T/citología
4.
J Immunol ; 180(10): 7009-18, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18453623

RESUMEN

Non-disrupted pieces of primary human lung tumor implanted into NOD-scid IL2Rgamma(null) mice consistently result in successful xenografts in which tissue architecture, including tumor-associated leukocytes, stromal fibroblasts, and tumor cells are preserved for prolonged periods with limited host-vs-graft interference. Human CD45(+) tumor-associated leukocytes within the xenograft are predominantly CD3(+) T cells with fewer CD138(+) plasma cells. The effector memory T cells that had been shown to be quiescent in human lung tumor microenvironments can be activated in situ as determined by the production of human IFN-gamma in response to exogenous IL-12. Plasma cells remain functional as evidenced by production of human Ig. Significant levels of human IFN-gamma and Ig were detected in sera from xenograft-bearing mice for up to 9 wk postengraftment. Tumor-associated T cells were found to migrate from the microenvironment of the xenograft to the lung, liver, and primarily the spleen. At 8 wk postengraftment, a significant portion of cells isolated from the mouse spleens were found to be human CD45(+) cells. The majority of CD45(+) cells were CD3(+) and expressed a phenotype consistent with an effector memory T cell, consisting of CD4(+) or CD8(+) T cells that were CD45RO(+), CD44(+), CD62L(-), and CD25(-). Following adoptive transfer into non-tumor bearing NOD-scid IL2Rgamma(null) mice, these human T cells were found to expand in the spleen, produce IFN-gamma, and maintain an effector memory phenotype. We conclude that the NOD-scid IL2Rgamma(null) tumor xenograft model provides an opportunity to study tumor and tumor-stromal cell interactions in situ for prolonged periods.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Memoria Inmunológica , Neoplasias Pulmonares/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunofenotipificación , Interferón gamma/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Receptores de Interleucina-2/metabolismo , Trasplante Heterólogo
5.
Artículo en Inglés | MEDLINE | ID: mdl-18220947

RESUMEN

Cytokine therapy can induce tumor regression in cancer patients but systemic administration of cytokines is accompanied with severe toxicity. Loco-regional delivery represents an effective and less toxic alternative to systemic injection. However; the requirement for frequent repeated injections of recombinant cytokine or the logistical difficulties associated with gene-modification have limited wide-spread use of loco-regional therapy. A simpler alternative local delivery strategy involves the use of controlled-release cytokine depot formulations. These formulations provide the advantage that physiological doses of cytokines are directly released to the tumor microenvironment in a sustained manner. Anti-tumor efficacy of IL-2; IL-12; GM-CSF or TNFalpha-encapsulated polymer microspheres has been evaluated in syngeneic murine and human tumor /SCID mouse xenograft models. A single intra-tumoral injection of these formulations; particularly that of IL-12 in combination with GM-CSF or TNFalpha; promoted the regression of established primary tumors; induced systemic anti-tumor T- and NK-cell responses and achieved complete eradication of disseminated disease. Cellular and molecular analysis of post-therapy tumor microenvironment demonstrated that treatment promoted the activation of tumor-associated T-effector/memory cells; the elimination of CD4+ CD25+ Foxp3+ T-suppressors and the de novo priming of tumor-specific CD8+ T-effector cells. Long-term monitoring of post-therapy tumors revealed that reversal of intra-tumoral immune suppression was transient and that T-suppressor cells rapidly re-infiltrated tumors. Repeated treatment resurrected anti-tumor activity; however, therapeutic efficacy declined with each treatment cycle. The observed loss of therapeutic efficacy was associated with a progressive intensification of the post-treatment T-suppressor cell rebound. In contrast; depletion of T-suppressor cells with low dose chemotherapy prior to each cycle of treatment resulted in a dramatic enhancement of long-term therapeutic efficacy leading to complete remissions. Clinical implications of these findings are discussed herein.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Citocinas/administración & dosificación , Neoplasias/terapia , Animales , Ensayos Clínicos como Asunto , Citocinas/inmunología , Preparaciones de Acción Retardada , Humanos , Neoplasias/inmunología
6.
Clin Immunol ; 118(1): 66-76, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16185929

RESUMEN

Protein antigens have been covalently linked randomly to surface proteins on immature dendritic cells (DC). This has been achieved under physiological conditions using a heterobifunctional reagent that couples antigens to free thiol groups expressed on DC surface proteins. This results in a significant increase in the amount of antigen that is bound to DC, and the antigen/membrane protein complexes that are formed are rapidly internalized. DC, loaded covalently with either beta-galactosidase (beta-gal) or a tumor-associated immunoglobulin (Ig) when injected into mice, induce a beta-gal- or Ig-specific T cell response, and a protective anti-tumor immunity for tumors expressing either beta-gal or the targeted Ig. This response is shown here to be significantly greater than that which is induced by DC that are loaded with these antigens via the conventional antigen pulse protocol. These results establish a novel, safe, and viable approach of enhancing the effectiveness of DC-based vaccination strategies for B cell lymphoma.


Asunto(s)
Linfocitos B/inmunología , Vacunas contra el Cáncer/inmunología , Carcinoma/inmunología , Neoplasias del Colon/inmunología , Células Dendríticas/inmunología , Inmunoglobulinas/farmacología , Glicoproteínas de Membrana/farmacología , Animales , Antígenos de Neoplasias/inmunología , Carcinoma/terapia , Línea Celular Tumoral , Neoplasias del Colon/terapia , Células Dendríticas/metabolismo , Células Dendríticas/trasplante , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoglobulinas/metabolismo , Activación de Linfocitos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Tasa de Supervivencia , Linfocitos T/citología , Linfocitos T/inmunología
7.
Cancer Immunol Immunother ; 54(10): 944-52, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15846492

RESUMEN

Previous studies by others using transplantable murine tumor models have demonstrated that the administration of antibodies that block CTLA-4 interaction with B7 can provoke the elimination of established tumors, and that the tumor suppression is mediated by T-cells and/or cells expressing NK1.1. Studies from our lab have established in a human/severe combined immunodeficient (SCID) mouse chimeric model that autologous peripheral blood leukocytes (PBL) can suppress the growth of tumor xenografts in a PBL dose-dependent fashion, and that this suppression is dependent upon the patient's T and NK cells. Using this human/mouse chimeric model, we sought to determine whether an antibody blockade of CTLA-4 would enhance the anti-tumor response of a patient's PBL. It was first important to determine whether the tumor suppression observed in the SCID model was dependent upon CD28/B7 co-stimulation. Blockade of B7 with a human CTLA-4-Ig fusion protein completely abrogated the lymphocyte-mediated tumor suppression, confirming in this model that tumor suppression is dependent upon a CD28/B7 co-stimulation. Using two different CTLA-4 specific monoclonal antibodies, we observed that CTLA-4 blockade significantly enhanced the human lymphocyte-mediated tumor suppression in mice co-engrafted with PBL and tumor cells. This enhancement was observed in both an allogeneic setting (in which the PBL were allogeneic with respect to the tumor) and an autologous setting (in which the PBL and tumor were from the same patient). These results sustain the notion that human anti-tumor immune response can be augmented (in vivo) by blocking the interaction between CTLA-4 and B7.


Asunto(s)
Antígenos de Diferenciación/inmunología , Carcinoma de Células Escamosas/prevención & control , Neoplasias Pulmonares/prevención & control , Linfocitos T/inmunología , Abatacept , Animales , Anticuerpos Monoclonales/farmacología , Antígenos/genética , Antígenos/inmunología , Antígenos CD/inmunología , Antígenos Ly , Antígenos de Superficie , Antígeno B7-1/inmunología , Antígeno B7-2 , Antígenos CD28/inmunología , Antígeno CTLA-4 , Carcinoma de Células Escamosas/inmunología , Células Dendríticas/inmunología , Humanos , Inmunoconjugados/farmacología , Células Asesinas Naturales/inmunología , Lectinas Tipo C , Neoplasias Pulmonares/inmunología , Activación de Linfocitos , Glicoproteínas de Membrana/inmunología , Ratones , Ratones SCID , Subfamilia B de Receptores Similares a Lectina de Células NK , Proteínas/genética , Proteínas/inmunología , Inmunodeficiencia Combinada Grave/inmunología , Trasplante Heterólogo
8.
Cancer Res ; 62(24): 7254-63, 2002 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-12499267

RESUMEN

Tumor cells, injected s.c., were maintained until spontaneous metastases to the lungs were established in all of the mice. Mice were then treated with a single dose of cytokine-encapsulated biodegradable microspheres injected directly into primary s.c. tumors to achieve a local and sustained release of interleukin 12 (IL-12), granulocyte-macrophage colony-stimulating factor (GM-CSF), or a combination of these cytokines to the tumor microenvironment. The s.c. tumors were surgically excised 6 days after microsphere injections, and the mice were monitored for recurrence of the primary tumor, survival, and progression of metastatic disease. Combined neoadjuvant treatment with IL-12 and GM-CSF microspheres was superior to all other treatments in reducing the recurrence of primary tumors, enhancing postoperative survival, and suppressing established metastatic disease. Long-term survival analysis demonstrated that intratumoral injection of IL-12 + GM-CSF-loaded microspheres resulted in the complete cure of disseminated disease in the majority of the animals. The addition of systemic low-dose IL-2 therapy to the treatment protocol resulted in the loss of the antitumor activity induced by IL-12 + GM-CSF treatment. In vivo lymphocyte subset depletions established that both T- and natural killer-cell subsets were required for the suppression of primary and metastatic tumors. Long-term, tumor-specific T-cell activity was demonstrated by immunohistochemical analysis of metastatic lesions, IFN-gamma enzyme-linked immunosorbent spot (ELISPOT) assays and tumor challenge studies. These results establish that neoadjuvant in situ tumor immunotherapy with IL-12 + GM-CSF microspheres induces both innate and adaptive antitumor immune responses resulting in the eradication of disseminated disease.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias del Colon/terapia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Inmunoterapia/métodos , Interleucina-12/administración & dosificación , Neoplasias Pulmonares/terapia , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/inmunología , Neoplasias del Colon/inmunología , Neoplasias del Colon/cirugía , Relación Dosis-Respuesta Inmunológica , Sinergismo Farmacológico , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/sangre , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Interferón gamma/biosíntesis , Interferón gamma/sangre , Interferón gamma/inmunología , Interleucina-12/sangre , Interleucina-12/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/cirugía , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Microesferas , Terapia Neoadyuvante , Trasplante de Neoplasias , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
9.
Cancer Res ; 62(9): 2611-7, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11980657

RESUMEN

A human/severe combined immunodeficient mouse chimeric model was used to demonstrate that peripheral blood leukocytes (PBLs) from a patient with lung cancer completely suppress the growth of an autologous tumor in a PBL dose-dependent fashion repeatedly and over a 4-year period. Suppression of the patient's tumor required CD4+ T cells, CD56+ natural killer cells, and CD14+ monocytes/macrophages, but was completely independent of CD8+ T cells. The CD4+ effector cells promoted tumor killing indirectly because direct tumor recognition and killing are precluded by the absence of MHC class I and II molecules on the tumor cells. Tumor suppression was found to require both human interleukin-12 (IL-12) and IFN-gamma, which were produced and released by the patient's monocytes and T cells, respectively. These results establish that human CD4+ T cells present in the peripheral blood of a patient with lung cancer are able to orchestrate cytokine-dependent killing of an autologous MHC-negative tumor indirectly and without codependence on CD8+ T cells. We conclude that human tumor suppression is achieved in vivo even in the absence of MHC molecules on tumor cells. This tumor suppression is mediated indirectly by cytokines produced by the patient's PBLs that ultimately initiate tumor killing via several, presently incompletely defined mechanisms.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Antígenos HLA/inmunología , Interferón gamma/inmunología , Interleucina-12/inmunología , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Linfocitos T Reguladores/inmunología , Animales , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/terapia , División Celular/inmunología , Femenino , Humanos , Inmunoterapia Adoptiva/métodos , Interferón gamma/farmacología , Receptores de Lipopolisacáridos/inmunología , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/terapia , Masculino , Ratones , Ratones SCID , Proteínas Recombinantes , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA