Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ann Rheum Dis ; 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38777379

RESUMEN

OBJECTIVE: Tissue-resident memory cells (Trm) are a subset of T cells residing persistently and long-term within specific tissues that contribute to persistent inflammation and tissue damage. We characterised the phenotype and function of Trm and the role of CD103 in primary Sjogren's syndrome (pSS). METHODS: In both pSS and non-pSS sicca syndrome patients, we examined Trm frequency, cytokine production in salivary glands (SG) and peripheral blood (PB). We also analysed Trm-related gene expression in SG biopsies through bulk and single-cell RNA sequencing (scRNAseq). Additionally, we investigated Trm properties in an immunisation-induced animal model of pSS (experimental SS, ESS) mouse model and assessed the effects of Trm inhibition via intraglandular anti-CD103 monoclonal antibody administration. RESULTS: Transcriptomic pSS SG showed an upregulation of genes associated with tissue recruitment and long-term survival of Trm cells, confirmed by a higher frequency of CD8+CD103+CD69+ cells in pSS SG, compared with non-specific sialadenitis (nSS). In SG, CD8+ CD103+ Trm contributed to the secretion of granzyme-B and interferon-γ, CD8+ Trm cells were localised within inflammatory infiltrates, where PD1+CD8+ T cells were also increased compared with nSS and MALT lymphoma. scRNAseq of PB and pSS SG T cells confirmed expression of CD69, ITGAE, GZMB, GZMK and HLA-DRB1 among CD3+CD8+ SG T cells. In the SG of ESS, CD8+CD69+CD103+ Trm producing Granzyme B progressively expanded. However, intraglandular blockade of CD103 in ESS reduced Trm, reduced glandular damage and improved salivary flow. CONCLUSIONS: CD103+CD8+Trm cells are expanded in the SG of pSS and ESS, participate in tissue inflammation and can be therapeutically targeted.

2.
Cancer Immunol Immunother ; 69(10): 1959-1972, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32388678

RESUMEN

Cancer vaccine development has proven challenging with the exception of some virally induced cancers for which prophylactic vaccines exist. Currently, there is only one FDA approved vaccine for the treatment of prostate cancer and as such prostate cancer continues to present a significant unmet medical need. In this study, we examine the effectiveness of a therapeutic cancer vaccine that combines the ISCOMATRIX™ adjuvant (ISCOMATRIX) with the Toll-like receptor 3 agonist, polyinosinic-polycytidylic acid (Poly I:C), and Flt3L, FMS-like tyrosine kinase 3 ligand. We employed the TRAMP-C1 (transgenic adenocarcinoma of the mouse prostate) model of prostate cancer and the self-protein mPAP (prostatic acid phosphatase) as the tumor antigen. ISCOMATRIX™-mPAP-Poly I:C-Flt3L was delivered in a therapeutic prime-boost regime that was consistently able to achieve complete tumor regression in 60% of animals treated and these tumor-free animals were protected upon rechallenge. Investigations into the underlying immunological mechanisms contributing to the effectiveness of this vaccine identified that both innate and adaptive responses are elicited and required. NK cells, CD4+ T cells and interferon-γ were all found to be critical for tumor control while tumor infiltrating CD8+ T cells became disabled by an immunosuppressive microenvironment. There is potential for broader application of this cancer vaccine, as we have been able to demonstrate effectiveness in two additional cancer models; melanoma (B16-OVA) and a model of B cell lymphoma (Eµ-myc-GFP-OVA).


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Colesterol/administración & dosificación , Melanoma Experimental/inmunología , Fosfolípidos/administración & dosificación , Neoplasias de la Próstata/inmunología , Saponinas/administración & dosificación , Animales , Apoptosis , Linfocitos T CD8-positivos/efectos de los fármacos , Proliferación Celular , Modelos Animales de Enfermedad , Combinación de Medicamentos , Humanos , Interferón gamma/metabolismo , Masculino , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Poli I-C/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Am J Pathol ; 186(1): 172-84, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26718978

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) is a regulator of neutrophil production, function, and survival. Herein, we investigated the role of G-CSF in a murine model of human uveitis-experimental autoimmune uveoretinitis. Experimental autoimmune uveoretinitis was dramatically reduced in G-CSF-deficient mice and in anti-G-CSF monoclonal antibody-treated, wild-type (WT) mice. Flow cytometric analysis of the ocular infiltrate in WT mice with experimental autoimmune uveoretinitis showed a mixed population, comprising neutrophils, macrophages, and T cells. The eyes of G-CSF-deficient and anti-G-CSF monoclonal antibody-treated WT mice had minimal neutrophil infiltrate, but no change in other myeloid-derived inflammatory cells. Antigen-specific T-cell responses were maintained, but the differentiation of pathogenic type 17 helper T cells in experimental autoimmune uveoretinitis was reduced with G-CSF deficiency. We show that G-CSF controls the ocular neutrophil infiltrate by modulating the expression of C-X-C chemokine receptors 2 and 4 on peripheral blood neutrophils, as well as actin polymerization and migration. These data reveal an integral role for G-CSF-driven neutrophil responses in ocular autoimmunity, operating within and outside of the bone marrow, and also identify G-CSF as a potential therapeutic target in the treatment of human uveoretinitis.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Factor Estimulante de Colonias de Granulocitos/inmunología , Neutrófilos/inmunología , Uveítis/inmunología , Animales , Enfermedades Autoinmunes/patología , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Uveítis/patología
4.
J Immunol ; 194(5): 2199-207, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25646304

RESUMEN

The development of therapeutic vaccines for treatment of established cancer has proven challenging. Cancer vaccines not only need to induce a robust tumor Ag-specific immune response but also need to overcome the tolerogenic and immunosuppressive microenvironments that exist within many solid cancers. ISCOMATRIX adjuvant (ISCOMATRIX) is able to induce both tumor Ag-specific cellular and Ab responses to protect mice against tumor challenge, but this is insufficient to result in regression of established solid tumors. In the current study, we have used B16-OVA melanoma, Panc-OVA pancreatic, and TRAMP-C1 prostate cancer mouse tumor models to test therapeutic efficacy of ISCOMATRIX vaccines combined with other immune modulators. The coadministration of an ISCOMATRIX vaccine with the TLR3 agonist, polyinosinic-polycytidylic acid, and TLR9 agonist, CpG, reduced tumor growth in all tumor models and the presence of ISCOMATRIX in the formulation was critical for the therapeutic efficacy of the vaccine. This vaccine combination induced a robust and multifunctional CD8(+) T cell response. Therapeutic protection required IFN-γ and CD8(+) T cells, whereas NK and CD4(+) T cells were found to be redundant. ISCOMATRIX vaccines combined with TLR3 and TLR9 agonists represent a promising cancer immunotherapy strategy.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Vacunas contra el Cáncer/administración & dosificación , Colesterol/administración & dosificación , Melanoma Experimental/terapia , Neoplasias Pancreáticas/terapia , Fosfolípidos/administración & dosificación , Neoplasias de la Próstata/terapia , Saponinas/administración & dosificación , Neoplasias Cutáneas/terapia , Adyuvantes Inmunológicos/administración & dosificación , Animales , Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Combinación de Medicamentos , Humanos , Inmunoterapia/métodos , Masculino , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/mortalidad , Ratones , Ratones Noqueados , Trasplante de Neoplasias , Oligodesoxirribonucleótidos/farmacología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/mortalidad , Poli I-C/farmacología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/mortalidad , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Análisis de Supervivencia , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 9/antagonistas & inhibidores , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología , Carga Tumoral/efectos de los fármacos
5.
J Immunol ; 192(7): 3259-68, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24610009

RESUMEN

Adjuvants are an essential component of modern vaccines and used for their ability to elicit immunity to coadministered Ags. Many adjuvants in clinical development are particulates, but how they drive innate and adaptive immune responses remains poorly understood. Studies have shown that a number of vaccine adjuvants activate inflammasome pathways in isolated APCs. However, the contribution of inflammasome activation to vaccine-mediated immunity in vivo remains controversial. In this study, we evaluated immune cell responses to the ISCOMATRIX adjuvant (IMX) in mice. Like other particulate vaccine adjuvants, IMX potently activated the NALP-3-ASC-Caspase-1 inflammasome in APCs, leading to IL-1ß and IL-18 production. The IL-18R pathway, but not IL-1R, was required for early innate and subsequent cellular immune responses to a model IMX vaccine. APCs directly exposed to IMX underwent an endosome-mediated cell-death response, which we propose initiates inflammatory events locally at the injection site. Importantly, both inflammasome-related and -unrelated pathways contributed to IL-18 dependence in vivo following IMX administration. TNF-α provided a physiological priming signal for inflammasome-dependent IL-18 production by APCs, which correlated with reduced vaccine-mediated immune cell responses in TNF-α- or TNFR-deficient mice. Taken together, our findings highlight an important disconnect between the mechanisms of vaccine adjuvant action in vitro versus in vivo.


Asunto(s)
Colesterol/inmunología , Inmunidad/inmunología , Inflamasomas/inmunología , Interleucina-18/inmunología , Fosfolípidos/inmunología , Saponinas/inmunología , Adenosina Trifosfato/inmunología , Adenosina Trifosfato/metabolismo , Adyuvantes Inmunológicos/farmacología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Western Blotting , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Colesterol/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Combinación de Medicamentos , Humanos , Inmunidad/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Lisosomas/efectos de los fármacos , Lisosomas/inmunología , Lisosomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Fosfolípidos/farmacología , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/inmunología , Saponinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
6.
J Immunol ; 192(11): 5031-8, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24760152

RESUMEN

High-dose i.v. Ig (IVIG) is used to treat various autoimmune and inflammatory diseases; however, the mechanism of action remains unclear. Based on the K/BxN serum transfer arthritis model in mice, IVIG suppression of inflammation has been attributed to a mechanism involving basophils and the binding of highly sialylated IgG Fc to DC-SIGN-expressing myeloid cells. The requirement for sialylation was examined in the collagen Ab-induced arthritis (CAbIA) and K/BxN serum transfer arthritis models in mice. High-dose IVIG (1-2 g/kg body weight) suppressed inflammatory arthritis when given prophylactically. The same doses were also effective in the CAbIA model when given subsequent to disease induction. In this therapeutic CAbIA model, the anti-inflammatory effect of IVIG was dependent on IgG Fc but not F(ab')2 fragments. Removal of sialic acid residues by neuraminidase had no impact on the anti-inflammatory activity of IVIG or Fc fragments. Treatment of mice with basophil-depleting mAbs did not abrogate the suppression of either CAbIA or K/BxN arthritis by IVIG. Our data confirm the therapeutic benefit of IVIG and IgG Fc in Ab-induced arthritis but fail to support the significance of sialylation and basophil involvement in the mechanism of action of IVIG therapy.


Asunto(s)
Artritis/inmunología , Artritis/prevención & control , Basófilos/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulinas Intravenosas/farmacología , Factores Inmunológicos/farmacología , Ácido N-Acetilneuramínico/inmunología , Animales , Artritis/patología , Basófilos/patología , Modelos Animales de Enfermedad , Inmunoglobulinas Intravenosas/inmunología , Factores Inmunológicos/inmunología , Masculino , Ratones , Ratones Endogámicos NOD
7.
Cancer Immunol Immunother ; 64(4): 507-18, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25662405

RESUMEN

Clinical outcomes from cancer vaccine trials in patients with advanced melanoma have so far been disappointing. This appears at least partially due to a state of immunosuppression in these patients induced by an expansion of regulatory cell populations including regulatory T cells (Tregs). We have previously demonstrated potent immunogenicity of the NY-ESO-1/ISCOMATRIX™ vaccine in patients with resected melanoma (study LUD99-08); however, the same vaccine induced only a few vaccine antigen-specific immune responses in patients with advanced disease (study LUD2002-013). Pre-clinical models suggest that the alkylating agent cyclophosphamide can enhance immune responses by depleting Tregs. Therefore, we have enrolled a second cohort of patients with advanced melanoma in the clinical trial LUD2002-013 to investigate whether pre-treatment with cyclophosphamide could improve the immunogenicity of the NY-ESO-1/ISCOMATRIX™ vaccine. The combination treatment led to a significant increase in vaccine-induced NY-ESO-1-specific CD4(+) T cell responses compared with the first trial cohort treated with vaccine alone. We could not detect a significant decline in regulatory T cells in peripheral blood of patients 14 days after cyclophosphamide administration, although a decline at an earlier time point cannot be excluded. Our observations support the inclusion of cyclophosphamide in combination trials with vaccines and other immune-modulatory agents.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antineoplásicos Alquilantes/administración & dosificación , Linfocitos T CD4-Positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Colesterol/inmunología , Ciclofosfamida/administración & dosificación , Melanoma/terapia , Proteínas de la Membrana/inmunología , Fosfolípidos/inmunología , Saponinas/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Vacunas contra el Cáncer/inmunología , Estudios de Cohortes , Terapia Combinada , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Femenino , Estudios de Seguimiento , Humanos , Metástasis Linfática , Masculino , Melanoma/inmunología , Melanoma/secundario , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Linfocitos T Reguladores/inmunología
8.
Eur J Immunol ; 43(2): 533-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23124877

RESUMEN

CD4(+) CD25(+) FoxP3(+) naturally occurring regulatory T (Treg) cells play a crucial role in the maintenance of immune tolerance and in preventing autoimmune pathology. Interventions that expand Treg cells are highly desirable, as they may offer novel treatment options in a variety of autoimmune and transplantation settings. Paralleling previous preclinical studies, we demonstrate here that administration of the hematopoietic growth factor Flt3L to human subjects increases the frequency and absolute number of Treg cells, and reduces the ratio of CD8(+) T cells to Treg cells in the peripheral blood. The increase in Treg cells was due to enhanced Treg-cell proliferation rather than release of Treg cells from the thymus. Further studies revealed that Flt3L-induced proliferation of Treg cells was an indirect effect that occurred via the interaction of Treg cells with the Flt3L-expanded pool of CD1c(+) myeloid dendritic cells. On the basis of these findings, Flt3L may represent a promising agent for promoting immune tolerance in a variety of clinical settings.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/inmunología , Proteínas de la Membrana/inmunología , Linfocitos T Reguladores/inmunología , Tirosina Quinasa 3 Similar a fms/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Procesos de Crecimiento Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Factores de Transcripción Forkhead/metabolismo , Humanos , Tolerancia Inmunológica/inmunología , Activación de Linfocitos/inmunología , Proteínas de la Membrana/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Linfocitos T Reguladores/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo
9.
J Immunol ; 187(1): 55-63, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21613613

RESUMEN

Cancer vaccines aim to induce CTL responses against tumors. Challenges for vaccine design are targeting Ag to dendritic cells (DCs) in vivo, facilitating cross-presentation, and conditioning the microenvironment for Th1 type immune responses. In this study, we report that ISCOM vaccines, which consist of ISCOMATRIX adjuvant and protein Ag, meet these challenges. Subcutaneous injection of an ISCOM vaccine in mice led to a substantial influx and activation of innate and adaptive immune effector cells in vaccine site-draining lymph nodes (VDLNs) as well as IFN-γ production by NK and NKT cells. Moreover, an ISCOM vaccine containing the model Ag OVA (OVA/ISCOM vaccine) was efficiently taken up by CD8α(+) DCs in VDLNs and induced their maturation and IL-12 production. Adoptive transfer of transgenic OT-I T cells revealed highly efficient cross-presentation of the OVA/ISCOM vaccine in vivo, whereas cross-presentation of soluble OVA was poor even at a 100-fold higher concentration. Cross-presenting activity was restricted to CD8α(+) DCs in VDLNs, whereas Langerin(+) DCs and CD8α(-) DCs were dispensable. Remarkably, compared with other adjuvant systems, the OVA/ISCOM vaccine induced a high frequency of OVA-specific CTLs capable of tumor cell killing in different tumor models. Thus, ISCOM vaccines combine potent immune activation with Ag delivery to CD8α(+) DCs in vivo for efficient induction of CTL responses.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Colesterol/administración & dosificación , Reactividad Cruzada/inmunología , Pruebas Inmunológicas de Citotoxicidad/métodos , Células Dendríticas/inmunología , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Fosfolípidos/administración & dosificación , Saponinas/administración & dosificación , Animales , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Células Cultivadas , Colesterol/inmunología , Células Dendríticas/metabolismo , Combinación de Medicamentos , Femenino , Técnicas de Sustitución del Gen , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Células T Asesinas Naturales/patología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Fosfolípidos/inmunología , Quillaja/inmunología , Saponinas/inmunología
10.
Immunol Cell Biol ; 90(5): 540-52, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21894173

RESUMEN

Generating a cytotoxic CD8(+) T-cell response that can eradicate malignant cells is the primary objective of cancer vaccine strategies. In this study we have characterized the innate and adaptive immune response to the ISCOMATRIX adjuvant, and the ability of vaccine antigens formulated with this adjuvant to promote antitumor immunity. ISCOMATRIX adjuvant led to a rapid innate immune cell response at the injection site, followed by the activation of natural killer and dendritic cells (DC) in regional draining lymph nodes. Strikingly, major histocompatibility complex (MHC) class I cross-presentation by CD8α(+) and CD8α(-) DCs was enhanced by up to 100-fold when antigen was formulated with ISCOMATRIX adjuvant. These coordinated features enabled efficient CD8(+) T-cell cross-priming, which exhibited prophylactic and therapeutic tumoricidal activity. The therapeutic efficacy of an ISCOMATRIX vaccine was further improved when co-administered with an anti-CD40 agonist antibody, suggesting that ISCOMATRIX-based vaccines may combine favorably with other immune modifiers in clinical development to treat cancer. Finally, we identified a requirement for the myeloid differentiation primary response gene 88 (MyD88) adapter protein for both innate and adaptive immune responses to ISCOMATRIX vaccines in vivo. Taken together, our findings support the utility of the ISCOMATRIX adjuvant for use in the development of novel vaccines, particularly those requiring strong CD8(+) T-cell immune responses, such as therapeutic cancer vaccines.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Colesterol/inmunología , Fosfolípidos/inmunología , Saponinas/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos de Neoplasias/inmunología , Antígenos CD40/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Vacunas contra el Cáncer/administración & dosificación , Colesterol/administración & dosificación , Reactividad Cruzada/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Combinación de Medicamentos , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Ovalbúmina/inmunología , Fosfolípidos/administración & dosificación , Receptor Cross-Talk/efectos de los fármacos , Saponinas/administración & dosificación , Transducción de Señal/efectos de los fármacos
11.
Blood ; 116(2): 218-25, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20430956

RESUMEN

The ability of dendritic cells (DCs) to cross-present protein tumor antigens to cytotoxic T lymphocytes (CTLs) underpins the success of therapeutic cancer vaccines. We studied cross-presentation of the cancer/testis antigen, NY-ESO-1, and the melanoma differentiation antigen, Melan-A by human DC subsets. Monocyte-derived DCs (MoDCs) efficiently cross-presented human leukocyte associated (HLA)-A2-restricted epitopes from either a formulated NY-ESO-1/ISCOMATRIX vaccine or when either antigen was mixed with ISCOMATRIX adjuvant. HLA-A2 epitope generation required endosomal acidification and was proteasome-independent for NY-ESO-1 and proteasome-dependent for Melan-A. Both MoDCs and CD1c(+) blood DCs cross-presented NY-ESO-1-specific HLA-A2(157-165)-, HLA-B7(60-72)-, and HLA-Cw3(92-100)-restricted epitopes when formulated as an NY-ESO-1/ISCOMATRIX vaccine, but this was limited when NY-ESO-1 and ISCOMATRIX adjuvant were added separately to the DC cultures. Finally, cross-presentation of NY-ESO-1(157-165)/HLA-A2, NY-ESO-1(60-72)/HLA-B7, and NY-ESO-1(92-100)/HLA-Cw3 epitopes was proteasome-dependent when formulated as immune complexes (ICs) but only proteasome-dependent for NY-ESO-1(60-72)/HLA-B7-restricted cross-presentation facilitated by ISCOMATRIX adjuvant. We demonstrate, for the first time, proteasome-dependent and independent cross-presentation of HLA-A-, B-, and C-restricted epitopes within the same full-length tumor antigen by human DCs. Our findings identify important differences in the capacities of human DC subsets to cross-present clinically relevant, full-length tumor antigens and how vaccine formulation impacts CTL responses in vivo.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Proteínas de Neoplasias/inmunología , Fragmentos de Péptidos/inmunología , Vacunas contra el Cáncer/inmunología , Colesterol/inmunología , Combinación de Medicamentos , Epítopos de Linfocito T/inmunología , Antígenos HLA-A/inmunología , Antígenos HLA-B/inmunología , Antígenos HLA-C/inmunología , Humanos , Activación de Linfocitos/inmunología , Antígeno MART-1 , Fosfolípidos/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Saponinas/inmunología
12.
Immunol Rev ; 222: 242-55, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18364006

RESUMEN

If one envisions dendritic cells (DCs) as nature's adjuvant, then it is easy to predict that they would be advantageous for cancer immunotherapy. Advances in culture processes that generate large numbers of purified and functionally mature DCs raised the possibility that DCs might be promising clinical agents to generate effective immune responses against cancer. The use of mature DCs as cellular vaccines was proposed to be superior to conventional strategies aimed at treating cancer, yet a phase III clinical trial in patients with melanoma demonstrated no increased benefit of DCs over standard therapy. Despite this and other apparent failures, we propose that DC-based therapy should not be discarded but rather reassessed. The heterogeneity of DCs and their interaction with other innate cells and regulatory and effector pathways must be clearly understood before the full therapeutic benefit of DCs are recognized. Several aspects of DC vaccination require optimization including the following: effective delivery of vaccines to DCs in lymphoid tissues; incorporation of components that induce appropriate DC activation; and facilitation of innate and adaptive interactions and reduction of regulatory T-cell networks or suppressive microenvironments that hinder the function of immune effectors. Application of this knowledge is resulting in encouraging new data in pre-clinical settings, where multiple arms of the immune system are targeted for cancer therapy.


Asunto(s)
Presentación de Antígeno , Vacunas contra el Cáncer , Células Dendríticas/inmunología , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/uso terapéutico , Ensayos Clínicos como Asunto , Células Dendríticas/patología , Humanos , Memoria Inmunológica , Inmunoterapia/tendencias , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Melanoma/inmunología , Melanoma/terapia , Ratones , Receptores de Interleucina-7/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
13.
Int J Cancer ; 128(4): 897-907, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20473889

RESUMEN

Vaccines based on immune stimulatory complexes (ISCOM) induce T-cell responses against tumor antigen (Ag). However, immune responses are impaired in pancreatic cancer patients. We investigated the efficacy of an ISCOM vaccine in a murine pancreatic carcinoma model. Panc02 cells expressing OVA as a model Ag were induced subcutaneously or orthotopically in the pancreas of C57BL/6 mice. Treatment consisted of an OVA containing ISCOM vaccine, either used alone or in combination with the TLR9 agonist CpG. The ISCOM vaccine effectively induced Ag-specific CTL capable of killing tumor cells. However, in mice with established tumors CTL induction by the vaccine was inefficient and did not affect tumor growth. Lack of efficacy correlated with increased numbers of Treg. Depletion of Treg with anti-CD25 mAb restored CTL induction and prolonged survival. Adding low-dose CpG to the ISCOM vaccine reduced Treg numbers, enhanced CTL responses and induced regression of pancreatic tumors in a CD8(+) T cell-dependent manner. Mice cured from the primary tumor mounted a memory T-cell response against wild-type Panc02 tumors, indicative of epitope spreading. Combining ISCOM vaccines with TLR agonists is a promising strategy for breaking tumor immune evasion and deserves further evaluation for the treatment of pancreatic carcinoma.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Modelos Animales de Enfermedad , Inmunoterapia , Oligodesoxirribonucleótidos/uso terapéutico , Neoplasias Pancreáticas/inmunología , Linfocitos T Reguladores/inmunología , Receptor Toll-Like 9/agonistas , Adenocarcinoma/inmunología , Adenocarcinoma/metabolismo , Adenocarcinoma/prevención & control , Animales , Complejo Antígeno-Anticuerpo/inmunología , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Evasión Inmune , Inmunización , Metástasis Linfática , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/genética , Ovalbúmina/inmunología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/prevención & control , Tasa de Supervivencia , Linfocitos T Citotóxicos/inmunología , Células Tumorales Cultivadas
14.
Cancer Immunol Immunother ; 60(11): 1625-37, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21698545

RESUMEN

BACKGROUND: NY-ESO-1 protein formulated in ISCOMATRIX™ results in CD4+, CD8+ T cell and antibody-mediated immunity. We evaluated persistence of immunity, relapse-free survival and tumour antigen expression upon relapse in patients vaccinated in an earlier trial. METHODS: Immunity was measured in 28 patients with resected NY-ESO-1-expressing tumours (melanoma 25, breast 3) 252-1,155 days (median = 681) after vaccination. In the earlier vaccination, trial patients received NY-ESO-1 with ISCOMATRIX™ adjuvant at three protein doses 10 µg, 30 µg or 100 µg (n = 14); 100 µg NY-ESO-1 protein (n = 8) or placebo (n = 6), together with 1 µg of intradermal (ID) NY-ESO-1 protein twice for DTH skin testing. Immune responses assessed in the current study included antibody titres, circulating NY-ESO-1-specific T cells and DTH reactivity 2 days after DTH skin testing with NY-ESO-1 protein (1 µg) or peptides (10 µg). Relapse-free survival was determined for 42 melanoma patients. On relapse NY-ESO-1 and HLA, class I was assessed by immunohistochemistry in 17. RESULTS: Persisting anti-NY-ESO-1 immunity was detected in 10/14 recipients who had previously received vaccine with ISCOMATRIX™ adjuvant. In contrast, immunity only persisted in 3/14 who received 100 µg un-adjuvanted NY-ESO-1 protein (3/8) or 2 µg DTH protein (0/6) P = 0.02. Hence, persisting NY-ESO-1 immunity was associated with prior adjuvant. Tumour NY-ESO-1 or HLA class I was downregulated in participants who relapsed suggesting immunoediting had occurred. CONCLUSION: Immunoediting suggests that a signal of anti-tumour activity was observed in high-risk resected melanoma patients vaccinated with NY-ESO-1/ISCOMATRIX™. This was associated with measurable persisting immunity in the majority of vaccinated subjects tested. A prospective randomised trial has been undertaken to confirm these results.


Asunto(s)
Antígenos de Neoplasias/administración & dosificación , Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/administración & dosificación , Colesterol/administración & dosificación , Melanoma/terapia , Proteínas de la Membrana/administración & dosificación , Fosfolípidos/administración & dosificación , Saponinas/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Adulto , Anciano , Secuencia de Aminoácidos , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/inmunología , Vacunas contra el Cáncer/inmunología , Colesterol/inmunología , Supervivencia sin Enfermedad , Regulación hacia Abajo , Combinación de Medicamentos , Hipersensibilidad a las Drogas/etiología , Hipersensibilidad a las Drogas/inmunología , Femenino , Humanos , Inmunohistoquímica , Masculino , Melanoma/inmunología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/inmunología , Persona de Mediana Edad , Datos de Secuencia Molecular , Fosfolípidos/inmunología , Estudios Prospectivos , Saponinas/inmunología , Piel/inmunología
15.
Blood ; 113(14): 3218-25, 2009 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-19141859

RESUMEN

Dendritic-cell (DC) and natural killer (NK)-cell interactions are critical in sculpting the adaptive immune response. However, the mechanisms by which DCs down-regulate NK-cell functions are not well understood. NK-cell function is inhibited by transforming growth factor beta (TGF-beta), but DCs do not appear to produce TGF-beta. We have previously shown that activated human DCs produce large amounts of activin-A, a TGF-beta superfamily member, which autoregulates DC function. The present report shows that NK-cells express type I and II activin receptors and that activin-A triggers NK-cell Smad 2/3 signaling. Furthermore, activin-A directly regulates NK cell functions by (1) down-regulating the T-box transcription factor T-bet and interferon gamma (IFN-gamma) but not perforin or granzyme mRNA; (2) suppressing NK-cell IFN-gamma production as potently as TGF-beta; and (3) suppressing NK-cell CD25 expression and proliferation and sculpting NK-cell cytokine and chemokine profiles. Interestingly, unlike TGF-beta, activin-A weakly down-regulates the NK-cell natural cytotoxicity receptors (NCRs) NKp30 and NKG2D but does not attenuate their cytotoxic function. These findings provide the first evidence for a novel immune regulatory role of activin-A during DC-mediated NK-cell regulation, highlighting the potential of antagonizing activin-A signaling in vivo to enhance NK cell-mediated immune functions and adaptive immunity.


Asunto(s)
Activinas/fisiología , Células Asesinas Naturales/fisiología , Activinas/metabolismo , Activinas/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Células Dendríticas/metabolismo , Regulación de la Expresión Génica , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Interferón gamma/metabolismo , Células Jurkat , Células K562 , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal/genética , Proteínas Smad/metabolismo , Proteínas Smad/fisiología
16.
J Immunol ; 182(3): 1253-9, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19155470

RESUMEN

Cancer vaccines aim to induce antitumor CTL responses, which require cross-presentation of tumor Ag to CTLs by dendritic cells (DCs). Adjuvants that facilitate cross-presentation of vaccine Ag are therefore key for inducing antitumor immunity. We previously reported that human DCs could not efficiently cross-present the full-length cancer/testis Ag NY-ESO-1 to CTL unless formulated as either an immune complex (NY-ESO-1/IC) or with ISCOMATRIX adjuvant. We now demonstrate that NY-ESO-1/ICs induce cross-presentation of HLA-A2- and HLA-Cw3-restricted epitopes via a proteasome-dependent pathway. In contrast, cross-presentation of NY-ESO-1/ISCOMATRIX vaccine was proteasome independent and required the cytosolic protease tripeptidyl peptidase II. Trafficking studies revealed that uptake of ICs and ISCOMATRIX vaccine by DCs occurred via endocytosis with delivery to lysosomes. Interestingly, ICs were retained in lysosomes, whereas ISCOMATRIX adjuvant induced rapid Ag translocation into the cytosol. Ag translocation was dependent on endosomal acidification and IL-4-driven differentiation of monocytes into DCs. This study demonstrates that Ag formulation determines Ag processing and supports a role for tripeptidyl peptidase II in cross-presentation of CTL epitopes restricted to diverse HLA alleles.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Colesterol/administración & dosificación , Reactividad Cruzada/inmunología , Citosol/inmunología , Células Dendríticas/inmunología , Proteínas de la Membrana/inmunología , Fosfolípidos/administración & dosificación , Saponinas/administración & dosificación , Serina Endopeptidasas/metabolismo , Aminopeptidasas , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/metabolismo , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/metabolismo , Diferenciación Celular/inmunología , Citosol/enzimología , Citosol/metabolismo , Células Dendríticas/enzimología , Células Dendríticas/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas , Combinación de Medicamentos , Endocitosis/inmunología , Endosomas/enzimología , Endosomas/inmunología , Endosomas/metabolismo , Humanos , Hidrólisis , Lisosomas/enzimología , Lisosomas/inmunología , Lisosomas/metabolismo , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/metabolismo , Células Progenitoras Mieloides/enzimología , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Transporte de Proteínas/inmunología , Quillaja/inmunología , Transducción de Señal/inmunología
17.
J Immunol ; 182(12): 7587-94, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494282

RESUMEN

We have cloned the mouse and human C-type lectin Clec12A, expressed both, and produced mAb recognizing both. Mouse Clec12A is highly expressed on splenic CD8(+) dendritic cells (DC) and plasmacytoid DC. A proportion of CD8(-)DC also expresses lower levels of Clec12A, as do monocytes, macrophages, and B cells. Human CLEC12A, like the mouse counterpart, is expressed on blood monocytes and DC, including pDC and BDCA-3(+)DC, the proposed equivalent of mouse CD8(+)DC. To determine whether Ag targeted to Clec12A could induce immune responses, mice were injected with a rat mAb recognizing Clec12A, or a control rat mAb, then production of anti-rat Ig was measured. Anti-Clec12A mAb alone produced only moderate responses, but these were amplified by coinjecting only small amounts of LPS as a DC activation agent. Furthermore, when OVA was conjugated to anti-Clec12A mAb, OVA-specific T cells were induced to proliferate. This Ag presentation to naive T cells was due to targeting conventional DC, because their ablation eliminated T cell activation. The potent Ab responses induced using microgram amounts of anti-Clec12A and minimal amounts of adjuvant demonstrate that this molecule can be used as an Ag-delivery target to enhance Ab responses to vaccines.


Asunto(s)
Formación de Anticuerpos/inmunología , Antígenos/inmunología , Células Dendríticas/inmunología , Lectinas Tipo C/inmunología , Receptores Mitogénicos/inmunología , Animales , Presentación de Antígeno/inmunología , Membrana Celular/inmunología , Células Cultivadas , Humanos , Leucocitos/inmunología , Ratones
18.
J Exp Med ; 198(10): 1495-506, 2003 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-14610045

RESUMEN

Extensive studies of mice deficient in one or several cytokine receptors have failed to support an indispensable role of cytokines in development of multiple blood cell lineages. Whereas B1 B cells and Igs are sustained at normal levels throughout life of mice deficient in IL-7, IL-7Ralpha, common cytokine receptor gamma chain, or flt3 ligand (FL), we report here that adult mice double deficient in IL-7Ralpha and FL completely lack visible LNs, conventional IgM+ B cells, IgA+ plasma cells, and B1 cells, and consequently produce no Igs. All stages of committed B cell progenitors are undetectable in FL-/- x IL-7Ralpha-/- BM that also lacks expression of the B cell commitment factor Pax5 and its direct target genes. Furthermore, in contrast to IL-7Ralpha-/- mice, FL-/- x IL-7Ralpha-/- mice also lack mature B cells and detectable committed B cell progenitors during fetal development. Thus, signaling through the cytokine tyrosine kinase receptor flt3 and IL-7Ralpha are indispensable for fetal and adult B cell development.


Asunto(s)
Linfocitos B/fisiología , Diferenciación Celular/fisiología , Proteínas de la Membrana/metabolismo , Receptores de Interleucina-7/metabolismo , Animales , Linfocitos B/citología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Ganglios Linfáticos Agregados/metabolismo , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/inmunología , Transducción de Señal/inmunología , Transducción de Señal/fisiología
19.
Blood ; 112(8): 3264-73, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18669894

RESUMEN

A novel dendritic cell (DC)-restricted molecule, Clec9A, was identified by gene expression profiling of mouse DC subtypes. Based on sequence similarity, a human ortholog was identified. Clec9A encodes a type II membrane protein with a single extracellular C-type lectin domain. Both the mouse Clec9A and human CLEC9A were cloned and expressed, and monoclonal antibodies (mAbs) against each were generated. Surface staining revealed that Clec9A was selective for mouse DCs and was restricted to the CD8(+) conventional DC and plasmacytoid DC subtypes. A subset of human blood DCs also expressed CLEC9A. A single injection of mice with a mAb against Clec9A, which targets antigens (Ags) to the DCs, produced a striking enhancement of antibody responses in the absence of added adjuvants or danger signals, even in mice lacking Toll-like receptor signaling pathways. Such targeting also enhanced CD4 and CD8 T-cell responses. Thus, Clec9A serves as a new marker to distinguish subtypes of both mouse and human DCs. Furthermore, targeting Ags to DCs with antibodies to Clec9A is a promising strategy to enhance the efficiency of vaccines, even in the absence of adjuvants.


Asunto(s)
Células Dendríticas/citología , Lectinas Tipo C/química , Secuencia de Aminoácidos , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Lectinas Tipo C/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Transducción de Señal , Vacunas/química , Vacunas/metabolismo
20.
Clin Cancer Res ; 15(6): 2166-73, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19276262

RESUMEN

PURPOSE: NY-ESO-1 is a highly immunogenic antigen expressed in a variety of malignancies, making it an excellent target for cancer vaccination. We recently developed a vaccine consisting of full-length recombinant NY-ESO-1 protein formulated with ISCOMATRIX adjuvant, which generated strong humoral and T-cell-mediated immune responses and seemed to reduce the risk of disease relapse in patients with fully resected melanoma. This study examines the clinical and immunologic efficacy of the same vaccine in patients with advanced metastatic melanoma. EXPERIMENTAL DESIGN: Delayed-type hypersensitivity responses, circulating NY-ESO-1-specific CD4(+) and CD8(+) T cells, and proportions of regulatory T cells (Treg) were assessed in patients. RESULTS: In contrast to patients with minimal residual disease, advanced melanoma patients showed no clinical responses to vaccination. Although strong antibody responses were mounted, the generation of delayed-type hypersensitivity responses was significantly impaired. The proportion of patients with circulating NY-ESO-1-specific CD4(+) T cells was also reduced, and although many patients had CD8(+) T cells specific to a broad range of NY-ESO-1 epitopes, the majority of these responses were preexisting. Tregs were enumerated in the blood by flow cytometric detection of cells with a CD4(+)CD25(+)FoxP3(+) and CD4(+)CD25(+)CD127(-) phenotype. Patients with advanced melanoma had a significantly higher proportion of circulating Treg compared with those with minimal residual disease. CONCLUSIONS: Our results point to a tumor-induced systemic immune suppression, showing a clear association between the stage of melanoma progression, the number of Treg in the blood, and the clinical and immunologic efficacy of the NY-ESO-1 ISCOMATRIX cancer vaccine.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Colesterol/administración & dosificación , Melanoma/terapia , Proteínas de la Membrana/inmunología , Fosfolípidos/administración & dosificación , Saponinas/administración & dosificación , Linfocitos T Reguladores/fisiología , Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos de Neoplasias/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Combinación de Medicamentos , Femenino , Humanos , Hipersensibilidad Tardía/etiología , Masculino , Proteínas de la Membrana/administración & dosificación , Persona de Mediana Edad , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA