Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Immunol ; 193(11): 5557-66, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25339667

RESUMEN

Adoptive immunotherapy using γδ T cells harnesses their natural role in tumor immunosurveillance. The efficacy of this approach is enhanced by aminobisphosphonates such as zoledronic acid and alendronic acid, both of which promote the accumulation of stimulatory phosphoantigens in target cells. However, the inefficient and nonselective uptake of these agents by tumor cells compromises the effective clinical exploitation of this principle. To overcome this, we have encapsulated aminobisphosphonates within liposomes. Expanded Vγ9Vδ2 T cells from patients and healthy donors displayed similar phenotype and destroyed autologous and immortalized ovarian tumor cells, following earlier pulsing with either free or liposome-encapsulated aminobisphosphonates. However, liposomal zoledronic acid proved highly toxic to SCID Beige mice. By contrast, the maximum tolerated dose of liposomal alendronic acid was 150-fold higher, rendering it much more suited to in vivo use. When injected into the peritoneal cavity, free and liposomal alendronic acid were both highly effective as sensitizing agents, enabling infused γδ T cells to promote the regression of established ovarian tumors by over one order of magnitude. Importantly however, liposomal alendronic acid proved markedly superior compared with free drug following i.v. delivery, exploiting the "enhanced permeability and retention effect" to render advanced tumors susceptible to γδ T cell-mediated shrinkage. Although folate targeting of liposomes enhanced the sensitization of folate receptor-α(+) ovarian tumor cells in vitro, this did not confer further therapeutic advantage in vivo. These findings support the development of an immunotherapeutic approach for ovarian and other tumors in which adoptively infused γδ T cells are targeted using liposomal alendronic acid.


Asunto(s)
Alendronato/administración & dosificación , Carcinoma/terapia , Inmunoterapia Adoptiva/métodos , Neoplasias Ováricas/terapia , Linfocitos T/efectos de los fármacos , Alendronato/química , Animales , Carcinoma/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Femenino , Humanos , Inmunización , Liposomas/química , Ratones , Ratones SCID , Neoplasias Ováricas/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/inmunología , Linfocitos T/trasplante , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Cancer Immunol Immunother ; 63(3): 215-24, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24297569

RESUMEN

The programmed death-1 (PD-1) pathway is important in the maintenance of peripheral tolerance and homeostasis through suppression of T cell receptor signaling. As such, it is employed by many tumors as a means of immune escape. We have investigated the role of this pathway in human ovarian cancer (OC) to assess its potential role as a diagnostic and/or prognostic marker and therapeutic target, following recent clinical trial success of antibody therapy directed at this pathway. We show programmed death ligand-1 (PD-L1) expression on monocytes in the ascites and blood of patients with malignant OC is strikingly higher than those with benign/borderline disease, with no overlap in the values between these groups. We characterize the regulation of this molecule and show a role of IL-10 present in ascitic fluid. Flow cytometric analysis of T cells present in the ascites and blood showed a correlation of PD-1 expression with malignant tumors versus benign/borderline, in a similar manner to PD-L1 expression on monocytes. Finally, we demonstrate functional links between PD-L1 expression on monocytes and OC tumor cells with suppression of T cell responses. Overall, we present data based on samples obtained from women with ovarian cancer, suggesting the PD-1 pathway may be used as a reliable diagnostic marker in OC, as well as a viable target for use with PD-1/PD-L1-directed antibody immunotherapy.


Asunto(s)
Antígeno B7-H1/metabolismo , Biomarcadores de Tumor/metabolismo , Terapia Molecular Dirigida/métodos , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Receptor de Muerte Celular Programada 1/metabolismo , Anticuerpos Bloqueadores/inmunología , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Carcinogénesis , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Terapia de Inmunosupresión , Interleucina-10/inmunología , Activación de Linfocitos , Monocitos/inmunología , Pronóstico , Linfocitos T/inmunología , Escape del Tumor
3.
J Immunol ; 191(5): 2437-45, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23898037

RESUMEN

Epithelial ovarian cancer (EOC) remains the most lethal gynecologic malignancy, underscoring the need for better therapies. Adoptive immunotherapy using genetically targeted T cells represents a promising new treatment for hematologic malignancies. However, solid tumors impose additional obstacles, including the lack of suitable targets for safe systemic therapy and the need to achieve effective T cell homing to sites of disease. Because EOC undergoes transcœlomic metastasis, both of these challenges may be circumvented by T cell administration to the peritoneal cavity. In this study, we describe such an immunotherapeutic approach for EOC, in which human T cells were targeted against the extended ErbB family, using a chimeric Ag receptor named T1E28z. T1E28z was coexpressed with a chimeric cytokine receptor named 4αß (combination termed T4), enabling the selective ex vivo expansion of engineered T cells using IL-4. Unlike control T cells, T4(+) T cells from healthy donors and patients with EOC were activated by and destroyed ErbB(+) EOC tumor cell lines and autologous tumor cultures. In vivo antitumor activity was demonstrated in mice bearing established luciferase-expressing SKOV-3 EOC xenografts. Tumor regression was accompanied by mild toxicity, manifested by weight loss. Although efficacy was transient, therapeutic response could be prolonged by repeated T cell administration. Furthermore, prior treatment with noncytotoxic doses of carboplatin sensitized SKOV-3 tumors to T4 immunotherapy, promoting enhanced disease regression using lower doses of T4(+) T cells. By combining these approaches, we demonstrate that repeated administration of carboplatin followed by T4(+) T cells achieved optimum therapeutic benefit in the absence of significant toxicity, even in mice with advanced tumor burdens.


Asunto(s)
Antineoplásicos/administración & dosificación , Carboplatino/administración & dosificación , Receptores ErbB/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias Glandulares y Epiteliales/terapia , Neoplasias Ováricas/terapia , Linfocitos T/inmunología , Animales , Carcinoma Epitelial de Ovario , Terapia Combinada , Quimioterapia/métodos , Femenino , Citometría de Flujo , Humanos , Ratones , Proteínas Recombinantes de Fusión/inmunología , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Reprod Sci ; 19(2): 123-34, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22138547

RESUMEN

The idea of using organ transplantation to solve quality-of-life issues was first introduced a century ago, with cornea transplants and thrusted before the world again in 1998, following a controversial hand transplant. Uterus transplantation (UTn) has been proposed as another quality-of-life transplant for the cure of permanent uterine factor infertility. In order to proceed in humans, a greater appreciation of the immunological mechanisms that underlie UTn is desirable. Allogeneic UTn (animal model) was first described by 2 studies in 1969. The first and only human UTn, performed in 2000, was an early attempt with limited use of animal model experiments prior to moving onto the human setting. Since then, work using rat, mouse, ovine, goat, and nonhuman primate models has demonstrated that the uterus is a very different but manageable organ immunologically compared to other transplanted organs. Therefore, specifically exploring immunological issues relating to UTn is a valuable and necessary part of the inevitable scientific process leading to successful human UTn.


Asunto(s)
Inmunología del Trasplante , Útero/trasplante , Animales , Modelos Animales de Enfermedad , Femenino , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Humanos , Infertilidad Femenina/etiología , Infertilidad Femenina/cirugía , Embarazo , Complicaciones del Embarazo/inmunología , Complicaciones del Embarazo/prevención & control , Enfermedades Uterinas/fisiopatología , Enfermedades Uterinas/cirugía , Útero/inmunología
5.
Blood ; 113(26): 6726-36, 2009 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-19273835

RESUMEN

Local inflammation during cutaneous leishmaniasis is accompanied by accumulation of CD11b(+) cells at the site of the infection. A functional role for these monocytic cells in local angiogenesis in leishmaniasis has not been described so far. Here, we show that CD11b(+) cells express high levels of the myeloid differentiation antigen carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1). In experimental cutaneous leishmaniasis in C57BL/6 wild-type (B6.WT) and B6.Ceacam1(-/-) mice, we found that only B6.Ceacam1(-/-) mice develop edemas and exhibit impairment of both hemangiogenesis and lymphangiogenesis. Because CEACAM1 expression correlates with functional angiogenesis, we further analyzed the role of the CD11b(+) population. In B6.Ceacam1(-/-) mice, we found systemic reduction of Ly-6C(high)/CD11b(high) monocyte precursors. To investigate whether CEACAM1(+) myeloid cells are causally related to efficient angiogenesis, we used reverse bone marrow transplants (BMTs) to restore CEACAM1(+) or CEACAM1(-) bone marrow in B6.Ceacam1(-/-) or B6.WT recipients, respectively. We found that angiogenesis was restored by CEACAM1(+) BMT only. In addition, we observed reduced morphogenic potential of inflammatory cells in Matrigel implants in CEACAM1(-) backgrounds or after systemic depletion of CD11b(high) macrophages. Taken together, we show for the first time that CEACAM1(+) myeloid cells are crucial for angiogenesis in inflammation.


Asunto(s)
Antígeno Carcinoembrionario/análisis , Inflamación/fisiopatología , Leishmaniasis Cutánea/fisiopatología , Células Mieloides/fisiología , Neovascularización Patológica/fisiopatología , Animales , Anticuerpos Antiprotozoarios/biosíntesis , Trasplante de Médula Ósea , Antígeno CD11b/análisis , Antígeno Carcinoembrionario/biosíntesis , Antígeno Carcinoembrionario/genética , Colágeno , Combinación de Medicamentos , Edema/etiología , Edema/patología , Glicoproteínas/biosíntesis , Inmunidad Celular , Implantes Experimentales , Inflamación/etiología , Inflamación/inmunología , Interferón gamma/biosíntesis , Laminina , Leishmania major/inmunología , Leishmaniasis Cutánea/complicaciones , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/patología , Vasos Linfáticos/metabolismo , Macrófagos/parasitología , Macrófagos/fisiología , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/química , Células Mieloides/clasificación , Neovascularización Patológica/patología , Proteoglicanos , Quimera por Radiación , Células TH1/inmunología
6.
J Immunol ; 182(2): 774-83, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19124720

RESUMEN

The biological role of Langerin+ dendritic cells (DCs) such as Langerhans cells and a subset of dermal DCs (dDCs) in adaptive immunity against cutaneous pathogens remains enigmatic. Thus, we analyzed the impact of Langerin+ DCs in adaptive T cell-mediated immunity toward Leishmania major parasites in a Lang-DTR mouse model that allows conditional diphtheria toxin (DT)-induced ablation of Langerin+ DCs in vivo. For the first time, infection experiments with DT-treated Lang-DTR mice revealed that proliferation of L. major-specific CD8+ T cells is significantly reduced during the early phase of the immune response following depletion of Langerin+ DCs. Consequently, the total number of activated CD8+ T cells within the draining lymph node and at the site of infection is diminished. Furthermore, we show that the impaired CD8+ T cell response is due to the absence of Langerin+ dDCs and not Langerhans cells. Nevertheless, the CD4+ T cell response is not altered and the infection is cleared as effectively in DT-treated Lang-DTR mice as in control mice. This clearly demonstrates that Langerin+ DCs are, in general, dispensable for an efficient adaptive immune response against L. major parasites. Thus, we propose a novel concept that, in the experimental model of leishmaniasis, priming of CD4+ T cells is mediated by Langerin- dDCs, whereas Langerin+ dDCs are involved in early priming of CD8+ T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/parasitología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/parasitología , Células Dendríticas/clasificación , Células Dendríticas/parasitología , Leishmaniasis Cutánea/inmunología , Activación de Linfocitos/inmunología , Animales , Antígenos de Superficie/genética , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Epidermis/inmunología , Epidermis/metabolismo , Epidermis/parasitología , Femenino , Técnicas de Sustitución del Gen/métodos , Inmunidad Innata , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Células de Langerhans/parasitología , Lectinas Tipo C/genética , Leishmania major/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/parasitología , Ganglios Linfáticos/patología , Activación de Linfocitos/genética , Lectinas de Unión a Manosa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...