Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Nat Med ; 12(2): 198-206, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16444264

RESUMEN

Little is known about the consequences of immune recognition of mutated gene products, despite their potential relevance to autoimmunity and tumor immunity. To identify mutations that induce immunity, here we have developed a systematic approach in which combinatorial DNA libraries encoding large numbers of random mutations in two syngeneic tyrosinase-related proteins are used to immunize black mice. We show that the libraries of mutated DNA induce autoimmune hypopigmentation and tumor immunity through cross-recognition of nonmutated gene products. Truncations are present in all immunogenic clones and are sufficient to elicit immunity to self, triggering recognition of normally silent epitopes. Immunity is further enhanced by specific amino acid substitutions that promote T helper cell responses. Thus, presentation of a vast repertoire of antigen variants to the immune system can enhance the generation of adaptive immune responses to self.


Asunto(s)
Autoantígenos/genética , Autoinmunidad/genética , Mutación , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/genética , Secuencia de Bases , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células COS , Chlorocebus aethiops , Reacciones Cruzadas , ADN Complementario/genética , Biblioteca de Genes , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/inmunología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Autotolerancia/genética , Transfección
2.
Cancer Cell ; 7(5): 403-5, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15894259

RESUMEN

Cancer is commonly described as a disease of genetic mutations. However, epidemiologic and clinical evidence points to the important but multifaceted role of the host. The immune system has something to say about cancer evolution through promotion of malignancy by inflammatory myeloid cells of the innate immune system. In a report in this issue of Cancer Cell, B cells are implicated as key players in the regulation of chronic inflammation that promotes early events in epithelial carcinogenesis. These are surprising observations, linking antibodies of the adaptive immune system to innate immune responses that drive epithelial carcinogenesis.


Asunto(s)
Linfocitos B/inmunología , Sistema Inmunológico/inmunología , Neoplasias Glandulares y Epiteliales/etiología , Animales , Enfermedad Crónica , Humanos , Inmunidad Innata/inmunología , Vigilancia Inmunológica/inmunología , Inflamación/complicaciones , Inflamación/inmunología , Macrófagos/inmunología , Ratones , Modelos Inmunológicos , Neoplasias Glandulares y Epiteliales/inmunología
3.
J Virol ; 85(20): 10814-25, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21835795

RESUMEN

Poxviruses are large DNA viruses that replicate in the cytoplasm of infected cells. Myxoma virus is a rabbit poxvirus that belongs to the Leporipoxvirus genus. It causes a lethal disease called myxomatosis in European rabbits but cannot sustain any detectable infection in nonlagomorphs. Vaccinia virus is a prototypal orthopoxvirus that was used as a vaccine to eradicate smallpox. Myxoma virus is nonpathogenic in mice, whereas systemic infection with vaccinia virus can be lethal even in immunocompetent mice. Plasmacytoid dendritic cells (pDCs) are potent type I interferon (IFN)-producing cells that play important roles in antiviral innate immunity. How poxviruses are sensed by pDCs to induce type I IFN production is not well understood. Here we report that infection of primary murine pDCs with myxoma virus, but not with vaccinia virus, induces IFN-α, IFN-ß, tumor necrosis factor (TNF), and interleukin-12p70 (IL-12p70) production. Using pDCs derived from genetic knockout mice, we show that the myxoma virus-induced innate immune response requires the endosomal DNA sensor TLR9 and its adaptor MyD88, transcription factors IRF5 and IRF7, and the type I IFN positive-feedback loop mediated by IFNAR1. It is independent of the cytoplasmic RNA sensing pathway mediated by the mitochondrial adaptor molecule MAVS, the TLR3 adaptor TRIF, or the transcription factor IRF3. Using pharmacological inhibitors, we demonstrate that myxoma virus-induced type I IFN and IL-12p70 production in murine pDCs is also dependent on phosphatidylinositol 3-kinase (PI3K) and Akt. Furthermore, our results reveal that the N-terminal Z-DNA/RNA binding domain of vaccinia virulence factor E3, which is missing in the orthologous M029 protein expressed by myxoma virus, plays an inhibitory role in poxvirus sensing and innate cytokine production by murine pDCs.


Asunto(s)
Células Dendríticas/inmunología , Factor 7 Regulador del Interferón/inmunología , Factores Reguladores del Interferón/inmunología , Interferón Tipo I/metabolismo , Factor 88 de Diferenciación Mieloide/inmunología , Myxoma virus/inmunología , Receptor Toll-Like 9/inmunología , Animales , Células Cultivadas , Femenino , Factor 7 Regulador del Interferón/metabolismo , Factores Reguladores del Interferón/metabolismo , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Interferón alfa y beta/inmunología , Receptor de Interferón alfa y beta/metabolismo , Receptor Toll-Like 9/metabolismo , Virus Vaccinia/inmunología
4.
Blood ; 115(22): 4384-92, 2010 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-20154220

RESUMEN

Cyclophosphamide (CTX), a commonly used chemotherapeutic agent can enhance immune responses. The ability of CTX to promote the proliferation of effector T cells and abrogate the function of regulatory T cells (Tregs) has been described. In this study, we examined the effects of CTX treatment on dendritic cell (DC) subsets and the subsequent outcome on the effector and suppressive arms of adaptive immunity. In secondary lymphoid tissues, tissue-derived migratory DCs (migratory DCs), lymphoid tissue-resident DCs (resident DCs), and plasmacytoid DCs (pDCs) are well described. CTX has profound and selective cytotoxic effects on CD8(+) resident DCs, but not skin-derived migratory DCs or pDCs in lymph nodes (LNs) and spleen, causing an imbalance among these DC subsets. CTX treatment increases the potency of DCs in antigen presentation and cytokine secretion, and partially inhibits the suppressor activity of Tregs. Adoptive transfer of CD8(+) DCs can reconstitute this population in regional draining LNs and abrogate the immune-enhancing effects of CTX in vivo. These findings demonstrate that CTX may improve immune responses by preferentially depleting CD8(+) lymphoid-resident DCs, which leads to diminished Treg suppression and enhanced effector T-cell function in vivo.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Ciclofosfamida/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Tejido Linfoide/efectos de los fármacos , Tejido Linfoide/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/efectos de los fármacos , Antígenos CD8/metabolismo , Células Dendríticas/clasificación , Femenino , Técnicas In Vitro , Ganglios Linfáticos/citología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Prueba de Cultivo Mixto de Linfocitos , Tejido Linfoide/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Inmunológicos , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Linfocitos T Reguladores/inmunología
5.
J Exp Med ; 201(12): 1881-4, 2005 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-15967819

RESUMEN

Around 700 BCE, a new military formation called the phalanx was established in ancient Greece: a tight column of heavy infantry carrying long spears, or pikes, used in a single prong of attack. Later, in the battle of Marathon described by Herodotus, the Greeks learned the advantages of multipronged attacks, a strategy still used in modern warfare. Is the immune system similar in its approach to combatting pathogens or tumors?


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Inmunidad Celular , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Antígenos CD , Antígenos de Diferenciación/inmunología , Antígeno CTLA-4 , Humanos , Proteínas Inmediatas-Precoces/inmunología , Fosfoproteínas/inmunología , Proteínas de la Matriz Viral/inmunología , Proteínas Virales/inmunología
6.
Blood ; 113(7): 1574-80, 2009 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-19011222

RESUMEN

Keratinocyte growth factor (KGF), which is given exogenously to allogeneic bone marrow transplantation (allo-BMT) recipients, supports thymic epithelial cells and increases thymic output of naive T cells. Here, we demonstrate that this improved T-cell reconstitution leads to enhanced responses to DNA plasmid tumor vaccination. Tumor-bearing mice treated with KGF and DNA vaccination have improved long-term survival and decreased tumor burden after allo-BMT. When assayed before vaccination, KGF-treated allo-BMT recipients have increased numbers of peripheral T cells, including CD8(+) T cells with vaccine-recognition potential. In response to vaccination, KGF-treated allo-BMT recipients, compared with control subjects, generate increased numbers of tumor-specific CD8(+) cells, as well as increased numbers of CD8(+) cells producing interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha). We also found unanticipated benefits to antitumor immunity with the administration of KGF. KGF-treated allo-BMT recipients have an improved ratio of T effector cells to regulatory T cells, a larger fraction of effector cells that display a central memory phenotype, and effector cells that are derived from a broader T-cell-receptor repertoire. In conclusion, our data suggest that KGF can function as a potent vaccine adjuvant after allo-BMT through its effects on posttransplantation T-cell reconstitution.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Vacunas contra el Cáncer/inmunología , Factor 7 de Crecimiento de Fibroblastos/farmacología , Timo/efectos de los fármacos , Vacunas de ADN/inmunología , Animales , Trasplante de Médula Ósea/métodos , Trasplante de Médula Ósea/mortalidad , Antígenos CD4/metabolismo , Linfocitos T CD8-positivos/citología , División Celular/efectos de los fármacos , División Celular/inmunología , Femenino , Factores de Transcripción Forkhead/metabolismo , Memoria Inmunológica/efectos de los fármacos , Memoria Inmunológica/inmunología , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Plásmidos , Tasa de Supervivencia , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Timo/citología , Timo/inmunología , Quimera por Trasplante , Trasplante Homólogo
7.
J Immunol ; 183(8): 4853-7, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19786544

RESUMEN

We report that OX40 stimulation drives all lineages of CD4 T cell development, including regulatory T cells (Tregs), and the plasticity of the response is dependant on local cytokines. In TGF-beta1-treated cultures, an OX40 agonist increased IFN-gamma and IL-4 production and diverted T cells from the Treg lineage. However, cytokine blockade in the context of OX40 stimulation promoted enhanced Treg accumulation. This observation was evident in naive mice, as OX40 engagement enhanced Treg proliferation and accumulation in vivo. Lastly, OX40 agonist administration influenced experimental autoimmune encephalomyelitis disease severity in opposing directions, depending on the timing of administration. Given during Ag priming, the OX40 agonist drove Treg expansion and inhibited disease, whereas given later it enhanced T cell effector cytokine production in the CNS and exacerbated disease. Hence, OX40 signaling can augment the accumulation of all CD4 T cell lineages; however, its accentuation of immune responses may have vastly different biologic outcomes depending upon the local cytokine milieu.


Asunto(s)
Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Receptores OX40/agonistas , Linfocitos T Reguladores/efectos de los fármacos , Traslado Adoptivo , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Antígenos CD28/efectos de los fármacos , Antígenos CD28/inmunología , Antígenos CD28/metabolismo , Complejo CD3/efectos de los fármacos , Complejo CD3/inmunología , Complejo CD3/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Interferón gamma/antagonistas & inhibidores , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interferón gamma/farmacología , Interleucina-4/antagonistas & inhibidores , Interleucina-4/inmunología , Interleucina-4/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores OX40/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/farmacología
8.
JCI Insight ; 6(20)2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34676831

RESUMEN

Only a subset of cancer patients responds to checkpoint blockade inhibition in the clinic. Strategies to overcome resistance are promising areas of investigation. Targeting glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) has shown efficacy in preclinical models, but GITR engagement is ineffective in controlling advanced, poorly immunogenic tumors, such as B16 melanoma, and has not yielded benefit in clinical trials. The alkylating agent cyclophosphamide (CTX) depletes regulatory T cells (Tregs), expands tumor-specific effector T cells (Teffs) via homeostatic proliferation, and induces immunogenic cell death. GITR agonism has an inhibitory effect on Tregs and activates Teffs. We therefore hypothesized that CTX and GITR agonism would promote effective antitumor immunity. Here we show that the combination of CTX and GITR agonism controlled tumor growth in clinically relevant mouse models. Mechanistically, we show that the combination therapy caused tumor cell death, clonal expansion of highly active CD8+ T cells, and depletion of Tregs by activation-induced cell death. Control of tumor growth was associated with the presence of an expanded population of highly activated, tumor-infiltrating, oligoclonal CD8+ T cells that led to a diminished TCR repertoire. Our studies show that the combination of CTX and GITR agonism is a rational chemoimmunotherapeutic approach that warrants further clinical investigation.


Asunto(s)
Ciclofosfamida/uso terapéutico , Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Inmunosupresores/uso terapéutico , Inmunoterapia/métodos , Linfocitos T Reguladores/inmunología , Animales , Ciclofosfamida/farmacología , Humanos , Inmunosupresores/farmacología , Ratones
9.
J Exp Med ; 200(6): 771-82, 2004 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-15381730

RESUMEN

Concomitant tumor immunity describes immune responses in a host with a progressive tumor that rejects the same tumor at a remote site. In this work, concomitant tumor immunity was investigated in mice bearing poorly immunogenic B16 melanoma. Progression of B16 tumors did not spontaneously elicit concomitant immunity. However, depletion of CD4(+) T cells in tumor-bearing mice resulted in CD8(+) T cell-mediated rejection of challenge tumors given on day 6. Concomitant immunity was also elicited by treatment with cyclophosphamide or DTA-1 monoclonal antibody against the glucocorticoid-induced tumor necrosis factor receptor. Immunity elicited by B16 melanoma cross-reacted with a distinct syngeneic melanoma, but not with nonmelanoma tumors. Furthermore, CD8(+) T cells from mice with concomitant immunity specifically responded to major histocompatibility complex class I-restricted epitopes of two melanocyte differentiation antigens. RAG1(-/-) mice adoptively transferred with CD8(+) and CD4(+) T cells lacking the CD4(+)CD25(+) compartment mounted robust concomitant immunity, which was suppressed by readdition of CD4(+)CD25(+) cells. Naturally occurring CD4(+)CD25(+) T cells efficiently suppressed concomitant immunity mediated by previously activated CD8(+) T cells, demonstrating that precursor regulatory T cells in naive hosts give rise to effective suppressors. These results show that regulatory T cells are the major regulators of concomitant tumor immunity against this weakly immunogenic tumor.


Asunto(s)
Melanoma Experimental/inmunología , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Antígenos de Diferenciación/inmunología , Ciclofosfamida/farmacología , Genes RAG-1 , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Interleucina-10/fisiología , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-2/análisis
10.
J Exp Med ; 200(2): 149-57, 2004 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-15249593

RESUMEN

Glucocorticoid-induced tumor necrosis factor receptor family-related gene (GITR) is a member of the tumor necrosis factor receptor (TNFR) family that is expressed at low levels on unstimulated T cells, B cells, and macrophages. Upon activation, CD4(+) and CD8(+) T cells up-regulate GITR expression, whereas immunoregulatory T cells constitutively express high levels of GITR. Here, we show that GITR may regulate alloreactive responses during graft-versus-host disease (GVHD) after allogeneic bone marrow transplantation (BMT). Using a BMT model with major histocompatibility complex class I and class II disparity, we demonstrate that GITR stimulation in vitro and in vivo enhances alloreactive CD8(+)CD25(-) T cell proliferation, whereas it decreases alloreactive CD4(+)CD25(-) proliferation. Allo-stimulated CD4(+)CD25(-) cells show increased apoptosis upon GITR stimulation that is dependent on the Fas-FasL pathway. Recipients of an allograft containing CD8(+)CD25(-) donor T cells had increased GVHD morbidity and mortality in the presence of GITR-activating antibody (Ab). Conversely, recipients of an allograft with CD4(+)CD25(-) T cells showed a significant decrease in GVHD when treated with a GITR-activating Ab. Our findings indicate that GITR has opposite effects on the regulation of alloreactive CD4(+) and CD8(+) T cells.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Enfermedad Injerto contra Huésped/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Apoptosis , Trasplante de Médula Ósea , División Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores de Interleucina-2/biosíntesis , Regulación hacia Arriba
11.
Proc Natl Acad Sci U S A ; 104(48): 19073-8, 2007 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-18045792

RESUMEN

The onconeural antigens appear to serve as tumor rejection antigens in the paraneoplastic neurologic disorders. Here, we used an unbiased peptide binding screen, followed by studies in HLA-A2.1 transgenic mice to identify naturally processed HLA-A2.1 restricted epitopes of the paraneoplastic cerebellar degeneration breast/ovarian cancer antigen cdr2. These mice were used to clone high-avidity cdr2-specific CD8(+) T cells that recognize human tumor cells presenting endogenously loaded MHC class I-cdr2 peptide. T cells with this specificity were detected in the peripheral blood of two HLA-A2.1(+) paraneoplastic cerebellar degeneration patients. We cloned T cell receptor (TCR) alpha and beta genes from cdr2-specific T cells; electroporation of RNA encoding this TCR turned nonreactive donor T cells into efficient killers of human cdr2-expressing tumor cells. Cloned cdr2-specific TCR genes provide a clinically relevant means for immunologic targeting of human gynecologic cancers.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas del Tejido Nervioso/inmunología , Neoplasias Ováricas/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Anciano , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/complicaciones , Línea Celular Tumoral , Citotoxicidad Inmunológica , Femenino , Antígeno HLA-A2/inmunología , Humanos , Inmunidad Innata , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Datos de Secuencia Molecular , Neoplasias Ováricas/complicaciones , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología
12.
J Clin Invest ; 116(5): 1382-90, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16614758

RESUMEN

T cells recognizing self antigens expressed by cancer cells are prevalent in the immune repertoire. However, activation of these autoreactive T cells is limited by weak signals that are incapable of fully priming naive T cells, creating a state of tolerance or ignorance. Even if T cell activation occurs, immunity can be further restricted by a dominant response directed at only a single epitope. Enhanced antigen presentation of multiple epitopes was investigated as a strategy to overcome these barriers. Specific point mutations that create altered peptide ligands were introduced into the gene encoding a nonimmunogenic tissue self antigen expressed by melanoma, tyrosinase-related protein-1 (Tyrp1). Deficient asparagine-linked glycosylation, which was caused by additional mutations, produced altered protein trafficking and fate that increased antigen processing. Immunization of mice with mutated Tyrp1 DNA elicited cross-reactive CD8(+) T cell responses against multiple nonmutated epitopes of syngeneic Tyrp1 and against melanoma cells. These multi-specific anti-Tyrp1 CD8(+) T cell responses led to rejection of poorly immunogenic melanoma and prolonged survival when immunization was started after tumor challenge. These studies demonstrate how rationally designed DNA vaccines directed against self antigens for enhanced antigen processing and presentation reveal novel self epitopes and elicit multi-specific T cell responses to nonimmunogenic, nonmutated self antigens, enhancing immunity against cancer self antigens.


Asunto(s)
Autoantígenos/química , Epítopos/química , Neoplasias/inmunología , Animales , Presentación de Antígeno , Asparagina/química , Linfocitos T CD8-positivos/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Oxidorreductasas/química , Oxidorreductasas/genética , Linfocitos T/inmunología
13.
Cancer Immun ; 9: 5, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19496531

RESUMEN

A differentiation antigen commonly expressed on melanoma cells, gp100 is the target of infiltrating T cells. We conducted a phase I randomized cross-over trial of melanoma patients with either xenogeneic (mouse) or human gp100 plasmid DNA injected intramuscularly at three dosages (100, 500 or 1,500 microg) every three weeks for three doses. After the first three injections, patients were then immunized three times with gp100 from the other species. Peripheral blood samples were analyzed at various time points following 10-day culture with gp100 peptides using multi-parametric flow cytometry. A total of 19 patients were enrolled, with 18 assessable for immune function and survival. 14 (74%) were male, with a median age of 56 years (range, 20-82). All patients had no evidence of disease; 10 (53%) had stage III disease, 3 each (16%) had stage IIB and IV disease, 2 (11%) had choroidal and 1 (5%) had anal mucosal involvement. With a median follow-up of 30 months, median progression-free survival (PFS) is 44 months. Median survival is not reached. There was no grade 3/4 toxicity; the most common grade 1/2 toxicity was an injection site reaction in 12 patients (63%, all grade 1). Five patients developed CD8+ cells binding gp100(280-288) HLA-A2-restricted tetramer. One patient had an increase in CD8+ IFN-gamma+ cells. This xenogeneic immunization strategy was safe and associated with minimal toxicity. There was also evidence of immune response.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/administración & dosificación , Melanoma/inmunología , Glicoproteínas de Membrana/inmunología , Neoplasias Cutáneas/inmunología , Linfocitos T/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/efectos adversos , Supervivencia sin Enfermedad , Estudios de Seguimiento , Antígeno HLA-A2 , Humanos , Inyecciones Intramusculares , Activación de Linfocitos , Masculino , Melanoma/terapia , Glicoproteínas de Membrana/genética , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Fragmentos de Péptidos , Péptidos , Neoplasias Cutáneas/terapia , Linfocitos T/inmunología , Linfocitos T/patología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/efectos adversos , Antígeno gp100 del Melanoma
14.
Cytotherapy ; 11(7): 912-22, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19903103

RESUMEN

BACKGROUND AIMS: Monitoring cellular immune responses is one prerequisite for the rational development of cancer vaccines. METHODS: We describe an extensive effort to optimize and validate quantitatively an in vitro T-cell culture method by determining the phenotype and function of both CD4(+) and CD8(+) T cells, including measurement of the phenotype markers CCR7, CD45RA, CD28 and CD27 and the functional markers interferon (IFN)-gamma, interleukin (IL)-2, macrophage inflammatory protein (MIP)-1beta, tumor necrosis factor (TNF)-alpha and CD107a. RESULTS: Autologous peripheral blood mononuclear cells (PBMC) were potent stimulators that expanded antigen (Ag)-specific CD8(+) T cells during short-term culture with the addition of IL-2 and IL-15 cytokines. Polyfunctional Ag-specific CD4(+) and CD8(+) T cells were detectable using this method. CONCLUSIONS: Our culture system represents a robust human T-cell culture protocol that permits phenotypic, quantitative and qualitative evaluation of vaccine-induced CD4(+) and CD8(+) T-cell responses.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Técnicas de Cultivo de Célula , Neoplasias/inmunología , Antígenos/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Vacunas contra el Cáncer , Células Cultivadas , Citocinas/metabolismo , Estudios de Factibilidad , Humanos , Neoplasias/terapia
15.
Mol Ther ; 16(4): 773-81, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18301399

RESUMEN

Successful approaches to tumor immunotherapy must overcome the physiological state of tolerance of the immune system to self-tumor antigens. Immunization with appropriate variants of syngeneic antigens can achieve this. However, improvements in vaccine design are needed for efficient cancer immunotherapy. Here we explore nine different chimeric vaccine designs, in which the antigen of interest is expressed as an in-frame fusion with polypeptides that impact antigen processing or presentation. In DNA immunization experiments in mice, three of nine fusions elevated relevant CD8(+) T-cell responses and tumor protection relative to an unfused melanoma antigen. These fusions were: Escherichia coli outer membrane protein A (OmpA), Pseudomonas aeruginosa exotoxin A, and VP22 protein of herpes simplex virus-1. The gains of immunogenicity conferred by the latter two are independent of epitope presentation by major histocompatibility complex class II (MHC II). This finding has positive implications for immunotherapy in individuals with CD4(+) T-cell deficiencies. We present evidence that antigen instability is not a sine qua non condition for immunogenicity. Experiments using two additional melanoma antigens identified different optimal fusion partners, thereby indicating that the benefits of fusion vectors remain antigen specific. Therefore large fusion vector panels such as those presented here can provide information to promote the successful advancement of gene-based vaccines.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Proteínas Recombinantes de Fusión/inmunología , Vacunas de ADN/inmunología , ADP Ribosa Transferasas/genética , ADP Ribosa Transferasas/inmunología , Animales , Presentación de Antígeno , Antígenos de Neoplasias/genética , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Linfocitos T CD8-positivos/inmunología , Células COS , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/uso terapéutico , Chlorocebus aethiops , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/inmunología , Exotoxinas/genética , Exotoxinas/inmunología , Femenino , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/genética , Vacunación , Vacunas de ADN/genética , Vacunas de ADN/uso terapéutico , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/inmunología , Factores de Virulencia/genética , Factores de Virulencia/inmunología , Exotoxina A de Pseudomonas aeruginosa
16.
Mol Ther ; 16(12): 2022-9, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18797450

RESUMEN

Granulocyte-macrophage colony-stimulating factor (GM-CSF) enhances immune responses by inducing proliferation, maturation, and migration of dendritic cells (DCs) as well as expansion and differentiation of B and T lymphocytes. The potency of DNA vaccines can be enhanced by the addition of DNA encoding cytokines, acting as molecular adjuvants. We conducted a phase I/II trial of human GM-CSF DNA in conjunction with a multipeptide vaccine (gp100 and tyrosinase) in stage III/IV melanoma patients. Nineteen human leukocyte antigen (HLA)-A*0201+ patients were treated. Three dose levels were studied: 100, 400, and 800 microg DNA/injection, administered subcutaneously every month with 500 microg of each peptide. In the dose-ranging study, three patients were treated at each dose level. The remaining patients were then treated at the highest dose. Most toxicities were grade 1 injection-site reactions. Eight patients (42%) developed CD8+ T-cell responses, defined by a > or =3 SD increase in baseline reactivity to tyrosinase or gp100 peptide in tetramer or intracellular cytokine staining (ICS) assays. There was no relationship between dose and T-cell response. Responding T cells had an effector memory cell phenotype. Polyfunctional T cells were also demonstrated. At a median of 31 months follow-up, median survival has not been reached. Human GM-CSF DNA was found to be a safe adjuvant.


Asunto(s)
Adyuvantes Inmunológicos/metabolismo , Vacunas contra el Cáncer/inmunología , Terapia Genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Melanoma/inmunología , Melanoma/terapia , Vacunas de ADN/inmunología , Adyuvantes Inmunológicos/genética , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Melanoma/patología , Estadificación de Neoplasias , Péptidos/efectos adversos , Péptidos/inmunología , Fenotipo , Proteínas Recombinantes , Vacunas de ADN/efectos adversos , Vacunas de ADN/genética , Vacunas de Subunidad/efectos adversos , Vacunas de Subunidad/inmunología
17.
Int J Cancer ; 123(12): 2832-9, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18798552

RESUMEN

During melanoma progression, patients develop anti-tumor immunity including the production of anti-tumor antibodies. Although the strategies developed by malignant cells to escape anti-tumor cellular immunity have been extensively investigated, little is known about tumor resistance to humoral immunity. The main effect of IgG antibodies is to activate the immune response by binding to host Fc gamma receptors (FcgammaR) expressed by immune cells. We previously reported in a limited study that some human metastatic melanoma cells ectopically express the FcgammaRIIB1, an inhibitory isoform of FcgammaR. By analyzing a large panel of different types of human primary and metastatic solid tumors, we report herein that expression of FcgammaRIIB is restricted to melanoma and is acquired during tumor progression. We show that FcgammaRIIB expression prevents the lysis of human metastatic melanoma cells by NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) in vitro, independently of the intracytoplasmic region of FcgammaRIIB. Using experimental mouse models, we demonstrate that expression of FcgammaRIIB protects B16F0 melanoma tumors from the ADCC induced by monoclonal and polyclonal anti-tumor IgG in vivo. Thus, our results identify FcgammaRIIB as a marker of human metastatic melanoma that impairs the tumor susceptibility to FcgammaR-dependent innate effector responses.


Asunto(s)
Biomarcadores de Tumor/análisis , Inmunoglobulina G/inmunología , Melanoma/inmunología , Receptores de IgG/análisis , Neoplasias Cutáneas/inmunología , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Melanoma/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Neoplasias Cutáneas/patología
18.
Adv Immunol ; 90: 215-41, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16730265

RESUMEN

A relationship between melanoma and vitiligo, a skin disorder characterized by the loss of melanocytes, has been postulated for many decades. In some cases, vitiligo is almost certainly a manifestation of autoimmune-mediated destruction of melanocytes. Melanocytes and melanoma cells share melanocyte differentiation antigens. Based on a number of observations, de novo vitiligo developing in patients with melanoma has been regarded as a sign of good prognosis. The immune system tolerates or ignores differentiation antigens because these antigens are self-derived. Therefore, immune tolerance or ignorance must be overcome to prime naive T and B cells to induce cancer immunity and autoimmunity against melanocyte differentiation antigens. Mouse models of concurrent melanoma and autoimmune vitiligo have revealed strategies to overcome immune ignorance or tolerance to melanocyte differentiation antigens: immunization with self-antigens as altered self (e.g., orthologues or mutated versions), expression in viral vectors, passive immunization with antibodies or T cells, incorporating potent adjuvants into active immunization, and blockade or removal of a downregulatory mechanism. Extensive investigations into the mechanisms that lead to tumor immunity and autoimmunity elicited by certain differentiation antigens have further revealed a variety of distinct cellular and molecular requirements, which are redundant and alternative.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Melanoma/inmunología , Vitíligo/inmunología , Animales , Antígenos de Diferenciación/administración & dosificación , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación/uso terapéutico , Autoanticuerpos/biosíntesis , Autoantígenos/administración & dosificación , Autoantígenos/inmunología , Autoantígenos/uso terapéutico , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/prevención & control , Humanos , Melanoma/diagnóstico , Melanoma/patología , Vacunación/métodos , Vacunación/tendencias , Vitíligo/diagnóstico , Vitíligo/patología
19.
Clin Cancer Res ; 13(10): 2977-85, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17504999

RESUMEN

PURPOSE: To determine the clinical toxicities and antibody response against sTn and tumor cells expressing sTn following immunization of high-risk breast cancer patients with clustered sTn-KLH [sTn(c)-KLH] conjugate plus QS-21. EXPERIMENTAL DESIGN: Twenty-seven patients with no evidence of disease and with a history of either stage IV no evidence of disease, rising tumor markers, stage II (>or=4 positive axillary nodes), or stage III disease received a total of five injections each during weeks 1, 2, 3, 7, and 19. Immunizations consisted of sTn(c)-KLH conjugate containing 30, 10, 3, or 1 microg sTn(c) plus 100 microg QS-21. Induction of IgM and IgG antibodies against synthetic sTn(c) and natural sTn on ovine submaxillary mucin were measured before and after therapy. Fluorescence-activated cell sorting analyses assessed reactivity of antibodies to LSC and MCF-7 tumor cells. RESULTS: The most common toxicities were transient local skin reactions at the injection site and mild flu-like symptoms. All patients developed significant IgM and IgG antibody titers against sTn(c). Antibody titers against ovine submaxillary mucin were usually of lower titers. IgM reactivity with LSC tumor cells was observed in 21 patients and with MCF-7 cells in 13 patients. There was minimal IgG reactivity with LSC cells. CONCLUSION: Immunization with sTn(c)-KLH conjugate plus QS-21 is well tolerated and immunogenic in high-risk breast cancer patients. Future trials will incorporate sTn(c) as a component of a multiple antigen vaccine.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Neoplasias de la Mama/prevención & control , Vacunas contra el Cáncer/uso terapéutico , Saponinas/uso terapéutico , Adyuvantes Inmunológicos/efectos adversos , Adulto , Anticuerpos/sangre , Neoplasias de la Mama/patología , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/inmunología , Femenino , Humanos , Inmunización , Inmunoglobulina M/sangre , Masculino , Persona de Mediana Edad , Riesgo , Saponinas/efectos adversos , Saponinas/inmunología
20.
Clin Cancer Res ; 13(20): 6195-203, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17947487

RESUMEN

PURPOSE: HER2/neu is frequently overexpressed in breast cancer. In a mouse model, vaccination with HER2/neu DNA elicits antibodies that confer partial protection against tumor challenge. EXPERIMENTAL DESIGN: To enhance antitumor immunity, we fused cDNA encoding Flt-3 ligand (FL) to the rat HER2/neu extracellular domain (neu), generating a chimeric FLneu molecule. FLneu and neu DNA vaccines were compared for immunogenicity and their ability to protect mice from tumor challenge. RESULTS: The neu vaccine generated a HER2/neu-specific antibody response. In contrast, vaccination with FLneu induced CD8+ T cells specific for HER2/neu but a negligible anti-HER2/neu antibody response. The switch from an antibody-mediated to T cell-mediated response was due to different intracellular localization of neu and FLneu. Although the neu protein was secreted, the FLneu protein was retained inside the cell, co-localizing with the endoplasmic reticulum, facilitating processing and presentation to T cells. The neu and FLneu vaccines individually conferred only weak tumor immunity. However, efficient tumor rejection was seen when neu and FLneu were combined, inducing both strong anti-HER2/neu-specific antibody and T cell responses. Adoptive transfer of both immune CD8+ T cells and immune sera from immunized mice was required to confer tumor immunity in naïve hosts. CONCLUSIONS: These results show that active induction of both humoral and cellular immunity to HER2/neu is required for efficient tumor protection, and that neither response alone is sufficient.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer , ADN/química , Proteínas de la Membrana/química , Neuraminidasa/química , Animales , Linfocitos T CD8-positivos/inmunología , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Humanos , Ligandos , Ratones , Microscopía Fluorescente , Trasplante de Neoplasias , Neuraminidasa/metabolismo , Estructura Terciaria de Proteína , Linfocitos T/metabolismo , Factores de Tiempo , Vacunas de ADN/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA