Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 615(7953): 687-696, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36356599

RESUMO

T cell receptors (TCRs) enable T cells to specifically recognize mutations in cancer cells1-3. Here we developed a clinical-grade approach based on CRISPR-Cas9 non-viral precision genome-editing to simultaneously knockout the two endogenous TCR genes TRAC (which encodes TCRα) and TRBC (which encodes TCRß). We also inserted into the TRAC locus two chains of a neoantigen-specific TCR (neoTCR) isolated from circulating T cells of patients. The neoTCRs were isolated using a personalized library of soluble predicted neoantigen-HLA capture reagents. Sixteen patients with different refractory solid cancers received up to three distinct neoTCR transgenic cell products. Each product expressed a patient-specific neoTCR and was administered in a cell-dose-escalation, first-in-human phase I clinical trial ( NCT03970382 ). One patient had grade 1 cytokine release syndrome and one patient had grade 3 encephalitis. All participants had the expected side effects from the lymphodepleting chemotherapy. Five patients had stable disease and the other eleven had disease progression as the best response on the therapy. neoTCR transgenic T cells were detected in tumour biopsy samples after infusion at frequencies higher than the native TCRs before infusion. This study demonstrates the feasibility of isolating and cloning multiple TCRs that recognize mutational neoantigens. Moreover, simultaneous knockout of the endogenous TCR and knock-in of neoTCRs using single-step, non-viral precision genome-editing are achieved. The manufacture of neoTCR engineered T cells at clinical grade, the safety of infusing up to three gene-edited neoTCR T cell products and the ability of the transgenic T cells to traffic to the tumours of patients are also demonstrated.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Edição de Genes , Neoplasias , Medicina de Precisão , Receptores de Antígenos de Linfócitos T , Linfócitos T , Transgenes , Humanos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Biópsia , Terapia Baseada em Transplante de Células e Tecidos/efeitos adversos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Síndrome da Liberação de Citocina/complicações , Progressão da Doença , Encefalite/complicações , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Genes Codificadores da Cadeia alfa de Receptores de Linfócitos T , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Mutação , Neoplasias/complicações , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/terapia , Segurança do Paciente , Medicina de Precisão/efeitos adversos , Medicina de Precisão/métodos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transgenes/genética , Antígenos HLA/imunologia , Sistemas CRISPR-Cas
2.
Bioconjug Chem ; 20(1): 111-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19102625

RESUMO

Polymeric carriers designed to encapsulate protein antigens have great potential for improving the efficacy of vaccines and immunotherapeutics for diseases such as cancer. We recently developed a carrier system based on polyacrylamide hydrogel microparticles cross-linked with acid-labile moieties. After being phagocytosed by antigen-presenting cells, the protein encapsulated within the carrier is released and processed for subsequent presentation of antigenic epitopes. To understand the impact of particle size on the activation of T-cells following uptake by antigen-presenting cells, particles with mean diameters of 3.5 microm and 35 nm encapsulating a model protein antigen were synthesized by emulsion and microemulsion based polymerization techniques, respectively. In vivo tests demonstrated that both sizes of particles were effective at stimulating the proliferation of T-cells and were capable of generating an antigen-specific cytotoxic T-cell response when coadministered with immunostimulatory DNA. Contrary to previous reports in the literature, our results suggest that there is no significant difference in the magnitude of T-cell activation for the two sizes of particles used in these experiments. This disparity in findings may be related to fundamental differences in material properties of the carriers used in these studies, such as the hydrophilicity of the polyacrylamide particles described here versus the hydrophobic nature of carriers investigated by other groups.


Assuntos
Antígenos/administração & dosagem , Portadores de Fármacos/química , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Linfócitos T/imunologia , Resinas Acrílicas/farmacologia , Animais , Humanos , Concentração de Íons de Hidrogênio , Tamanho da Partícula , Vacinas/química
3.
Methods Mol Biol ; 380: 457-68, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17876112

RESUMO

Dendritic cells (DC) are extremely potent antigen-presenting cells, which can prime both naive CD4+ and CD8+ T lymphocytes. In their immature state, DC continuously sample and process antigens from the surrounding environment, but only mature DC express sufficient levels of costimulatory molecules to activate naive T cells. DC present in tumors are functionally immature owing to the immunosuppressive actions of tumor-derived factors and regulatory T cells, and such immature DC promote immune tolerance to the tumor. Recent studies from animal models suggest that Toll-like receptor (TLR) agonists such as CpG can reverse the tolerogenic state of tumoral DC. Strategies that allow DC to gain access to both tumor antigens and TLR agonists, in situ, can overcome tumor tolerance leading to the induction of potent systemic antitumor immunity.


Assuntos
Antígenos de Neoplasias/imunologia , Células Dendríticas/imunologia , Tolerância a Antígenos Próprios/imunologia , Animais , Vacinas Anticâncer/imunologia , Humanos , Melanoma Experimental/imunologia , Melanoma Experimental/terapia
4.
Exp Hematol ; 33(2): 173-81, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15676211

RESUMO

OBJECTIVE: The aim of this study was to determine the lineage origin of interferon-alpha-producing cells (IPCs), also called plasmacytoid dendritic cells, in mice by evaluating the ability of common lymphoid (CLP) and myeloid (CMP) progenitors to give rise to IPCs. MATERIALS AND METHODS: Sublethally irradiated C57Bl/6 mice were intravenously transplanted with rigorously purified lymphoid and myeloid progenitors from a congenic mouse strain. At various time points posttransplantation mice were analyzed for donor-derived cells by flow cytometry. The developmental potential of all progenitor populations was also tested in in vitro cultures. In addition, in vitro and in vivo derived IPCs were functionally assessed for their interferon-alpha production after virus challenge. RESULTS: Transplantation of 1 x 10(4) common myeloid progenitors, 1 x 10(4) common lymphoid progenitors or 2.5 x 10(4) granulocyte/macrophage progenitors all led to the generation of IPCs within 2 to 3 weeks. In general, IPC reconstitution in spleen and liver by CMPs was more efficient than by CLP. Adding Flt3L alone to in vitro cultures was sufficient to support the development of IPCs from myeloid progenitors whereas CLPs required additional survival factors provided either by stroma cells or by introduction of transgenic Bcl-2. Both myeloid- and lymphoid-derived IPC were indistinguishable by function, gene expression, and morphology. CONCLUSION: Surprisingly, our results clearly show that murine IPCs differentiate from both lineages but are mainly of myeloid origin. These results extend to IPCs the observation made originally in classical dendritic cells that cellular expression of so called lineage markers does not correlate with lineal origin.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/imunologia , Células-Tronco Hematopoéticas/citologia , Interferon-alfa/genética , Interleucina-2/genética , Transplante de Células-Tronco , Animais , Antígenos CD/análise , Diferenciação Celular , Células-Tronco Hematopoéticas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Condicionamento Pré-Transplante , Irradiação Corporal Total
5.
Oncogene ; 21(10): 1571-9, 2002 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-11896586

RESUMO

Gfi-1 is a nuclear zinc finger protein with the activity of a transcriptional repressor and the ability to predispose for the development of T-cell lymphoma when expressed constitutively at high levels. Whereas thymic T-cell precursors express endogenous Gfi-1, mature peripheral T-cells lack Gfi-1 but upregulate its expression transiently after antigenic stimulation and activation of Erk1/2 demonstrating a role of Gfi-1 in T-cell activation. Here we show that constitutive expression of Gfi-1 accelerates S phase entry of primary, resting T-cells upon antigenic stimulation. In addition, high level Gfi-1 expression inhibits phorbol ester induced G1 arrest and activation induced cell death in Jurkat T-cells. We demonstrate that these effects of Gfi-1 concur with lower absolute levels and hyperphosphorylation of the pocket protein pRb. Moreover, phorbol ester induced expression of the negative cell cycle regulator p21(WAF1) is blocked in the presence of Gfi-1. These findings suggest that Gfi-1 contributes to T-cell lymphomagenesis by overriding a late G1 cell cycle checkpoint which controls activation induced death and S phase entry of T-cells.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/metabolismo , Ativação Linfocitária , Linfócitos T/imunologia , Fatores de Transcrição , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígenos CD2/genética , Ciclo Celular , Morte Celular , Células Cultivadas , Proteínas de Ligação a DNA/genética , Humanos , Células Jurkat , Cinética , Lectinas Tipo C , Camundongos , Camundongos Transgênicos , Proteínas Oncogênicas/biossíntese , RNA Mensageiro/biossíntese , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica , Regulação para Cima
6.
Ann N Y Acad Sci ; 1058: 96-104, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16394129

RESUMO

Dendritic cells (DC) are the most potent antigen-presenting cells known, capable of priming both naïve CD4 and CD8 T lymphocytes. In their immature state DC are especially efficient at ingesting dying cells and processing their antigens while in their mature state DC express high levels of both MHC class I and II antigens as well as a variety of costimulatory molecules needed to induce a primary T cell response. In addition to stimulating adaptive T cell responses, DC can stimulate NK cells and B cells. Their dual role in innate and adaptive immunity led us to explore their potential utility in tumor immunotherapy. The results of clinical trials in which autologous DC loaded with tumor antigen are administered to tumor-bearing patients have been promising, but overall immunologic potency and clinical efficacy have been unsatisfactory. It has become clear that more potent and more efficient DC based immunotherapies are needed, preferably based on in vivo targeting of DC rather than in vitro manipulation followed by systemic administration of the cells. Recent studies in animal models suggest that DC can be loaded with antigen and activated in vivo in a manner that results in potent antitumor immunity. Two approaches to DC targeting are described in this review, both of which have been shown to overcome immune tolerance to tumors and induce dramatic tumor regression.


Assuntos
Células Dendríticas/citologia , Imunoterapia/métodos , Animais , Antígenos de Neoplasias/metabolismo , Linfócitos B/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Ilhas de CpG , Células Dendríticas/metabolismo , Humanos , Tolerância Imunológica , Células Matadoras Naturais/metabolismo , Proteínas de Membrana , Modelos Biológicos
7.
Mol Pharm ; 6(4): 1160-9, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19415922

RESUMO

Protein-based vaccines have been explored as a safer alternative to traditional weakened or killed whole organism based vaccination strategies and have been investigated for their ability to activate the immune system against certain cancers. For optimal stimulation of T lymphocytes, protein-based vaccines should deliver protein antigens to antigen presenting cells in the context of appropriate immunostimulatory signals, thus mimicking actual pathogens. In this report, we describe the synthesis, characterization, and biological evaluation of immunostimulatory acid-degradable microparticles, which are suitable delivery vehicles for use in protein-based vaccines and cancer immunotherapy. Using a 3' conjugation strategy, we optimized the attachment of immunostimulatory CpG DNA to our vaccine carriers and demonstrated that under acidic conditions similar to those found in endosomal compartments, these new particles were capable of simultaneously releasing a model protein antigen and a CpG DNA adjuvant. We found in an in vivo cytotoxicity assay that the co-encapsulation of ovalbumin, a model antigen, and immunostimulatory agent in the same particle led to superior cytotoxic T lymphocyte activity compared to particles coadministered with adjuvant in an unbound form. In addition, we investigated the ability of these acid-degradable particles to induce protective immunity in the MO5 murine melanoma model and found that they were effective until tumor escape, which appeared to result from a loss of antigen expression by the cancer cells due to in vivo selection pressure.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antineoplásicos/administração & dosagem , Vacinas Anticâncer/uso terapêutico , Melanoma Experimental/terapia , Oligodesoxirribonucleotídeos/administração & dosagem , Ovalbumina/administração & dosagem , Adjuvantes Imunológicos/farmacocinética , Adjuvantes Imunológicos/farmacologia , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Citocinas/metabolismo , Sistemas de Liberação de Medicamentos , Feminino , Ativação Linfocitária , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas , Oligodesoxirribonucleotídeos/farmacocinética , Oligodesoxirribonucleotídeos/farmacologia , Ovalbumina/farmacocinética , Ovalbumina/farmacologia , Taxa de Sobrevida , Distribuição Tecidual
8.
Bioconjug Chem ; 18(1): 77-83, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17226959

RESUMO

The development of multicomponent biotherapeutic carriers is an important challenge in the field of drug delivery, particularly in the area of protein-based vaccines. While the delivery of protein antigens to antigen presenting cells (APCs) is crucial for this type of vaccination, the incorporation of additional adjuvants may be just as important in order to generate more potent immune responses. This article presents the synthesis and biological evaluation of carrier particles that both deliver a protein payload to APCs and display receptor ligands for the enhancement of APC immunostimulation. Particles displaying CpG oligonucleotide ligands for Toll-like receptor 9 were synthesized. The addition of CpG DNA to the particles led to a 45-fold increase in the secretion of interleukin-12, a cytokine that aids in T-cell activation, and a significant increase in the expression of costimulatory molecules by APCs. Moreover, vaccination with particles containing both ovalbumin (OVA) and CpG DNA induced a superior OVA-specific CD8 T-cell response in vivo, as measured by increased OVA-specific CD8 T-cell proliferation, secretion of the proinflammatory cytokine IFN-gamma, and the induction of OVA-specific cytotoxicity.


Assuntos
Antígenos/química , Antígenos/imunologia , Linfócitos T CD8-Positivos/imunologia , Oligodesoxirribonucleotídeos/imunologia , Ovalbumina/química , Ovalbumina/imunologia , Vacinas/imunologia , Animais , Biomarcadores , Proliferação de Células , Células Cultivadas , Células Dendríticas/imunologia , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Oligodesoxirribonucleotídeos/síntese química , Oligodesoxirribonucleotídeos/química
9.
Blood ; 107(4): 1383-90, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16263793

RESUMO

Langerhans cells (LCs) are antigen-presenting cells (APCs) residing in the epidermis that play a major role in skin immunity. Our earlier studies showed that when skin is inflamed LCs are replaced by bone marrow-derived progenitor cells, while during steady-state conditions LCs are able to self-renew in the skin. Identification of the LC progenitors in bone marrow would represent a critical step toward identifying the factors that regulate LC generation as well as their trafficking to the skin. To determine LC lineage origin, we reconstituted lethally irradiated CD45.2 mice with rigorously purified lymphoid and myeloid progenitors from CD45.1 congenic mice. Twenty-four hours later, we exposed the mice to UV light to deplete resident LCs and induce their replacement by progenitors. Reconstitution with common myeloid progenitors (CMPs), common lymphoid progenitors (CLPs), granulocyte-macrophage progenitors (GMPs), or early thymic progenitors led to LC generation within 2 to 3 weeks. CMPs were at least 20 times more efficient at generating LCs than CLPs. LCs from both lineages were derived almost entirely from fetal liver kinase-2+ (Flk-2+) progenitors, displayed typical dendritic-cell (DC) morphology, and showed long-term persistence in the skin. These results indicate that LCs are derived mainly from myeloid progenitors and are dependent on Flt3-ligand for their development.


Assuntos
Células de Langerhans/citologia , Células de Langerhans/imunologia , Células-Tronco/citologia , Tirosina Quinase 3 Semelhante a fms/fisiologia , Animais , Células Apresentadoras de Antígenos/fisiologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Diferenciação Celular , Primers do DNA , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/imunologia , Transplante de Células-Tronco , Tirosina Quinase 3 Semelhante a fms/genética
10.
Int Immunol ; 17(5): 539-47, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15778289

RESUMO

Conjugates of peptide antigens with antibodies specifically recognizing surface molecules on dendritic cells (DC) represent an attractive approach to target antigens to antigen-presenting cells (APC) for the induction of specific T cell responses. The present study evaluates the potential of M-DC8(+) DC, a sub-population of professional APC in the blood, for an antibody-based vaccination strategy. We prepared, by chemical cross-linking, conjugates of peptide model antigens with antibodies directed against different cell surface molecules of DC. Antigen-peptide conjugates using an anti-CD16 (FcgammaRIII) antibody were most potent in inducing in vitro activation of a specific CD4(+) T cell response. They were at least 300 times more efficient than two other antibody-antigen conjugates and approximately 500 times more efficient than unconjugated antigen peptides. Our data demonstrate that specific antigen targeting via CD16 on M-DC8(+) DC is a promising vaccination approach for the efficient induction of specific CD4(+) T cell responses ex vivo, and perhaps in vivo.


Assuntos
Apresentação de Antígeno , Células Dendríticas/imunologia , Imunoconjugados/imunologia , Receptores de IgG/imunologia , Anticorpos/imunologia , Antígenos/imunologia , Antígenos CD/imunologia , Linfócitos T CD4-Positivos/imunologia , Apresentação Cruzada , Células Dendríticas/classificação , Células Dendríticas/citologia , Humanos , Leucócitos Mononucleares/citologia , Ativação Linfocitária , Toxina Tetânica/imunologia , Vacinas
11.
J Immunol ; 170(10): 5089-94, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12734354

RESUMO

Recently, a new class of human dendritic cell (DC) precursors has been described in the peripheral blood recognized by the mAb M-DC8. These cells represent approximately 1% of PBMC and acquire several characteristics of myeloid DC upon in vitro culture. In this report we show that M-DC8(+) monocytes secrete in response to LPS >10 times the amount of TNF-alpha as M-DC8(-) monocytes, but produce significantly less IL-10. Consistent with a role in inflammatory responses, we found that M-DC8(+) cells localized in the T cell area of inflamed human tonsils and in the subepithelial dome region of Peyer's patches. In patients with active Crohn's disease, abundant M-DC8(+) cells were detectable in inflamed ileal mucosa, which were entirely depleted after systemic steroid treatment. Our results indicate that M-DC8(+) cells are cells of DC phenotype in inflamed mucosa-associated lymphoid tissue that may contribute to the high level of TNF-alpha production in Crohn's disease. We infer that selective elimination of M-DC8(+) cells in inflammatory diseases has therapeutic potential.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Tecido Linfoide/imunologia , Mucosa/imunologia , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Anti-Inflamatórios/uso terapêutico , Anticorpos Monoclonais/análise , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Infecções Bacterianas/imunologia , Infecções Bacterianas/patologia , Células Cultivadas , Criança , Doença de Crohn/tratamento farmacológico , Doença de Crohn/imunologia , Doença de Crohn/patologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/patologia , Dexametasona/farmacologia , Humanos , Íleo/efeitos dos fármacos , Íleo/imunologia , Íleo/patologia , Imuno-Histoquímica , Inflamação/imunologia , Inflamação/patologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Contagem de Leucócitos , Tecido Linfoide/metabolismo , Tecido Linfoide/microbiologia , Tecido Linfoide/patologia , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Mucosa/metabolismo , Mucosa/microbiologia , Mucosa/patologia , Tonsila Palatina/imunologia , Tonsila Palatina/patologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/patologia , Prednisona/uso terapêutico , Sepse/imunologia , Sepse/patologia , Linfócitos T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA