Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Clin Transl Immunology ; 13(2): e1490, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38375330

RESUMO

Objectives: Systemic inflammatory response syndrome (SIRS) is a frequent complication of cardiopulmonary bypass (CPB). SIRS is associated with significant morbidity and mortality, but its pathogenesis remains incompletely understood, and as a result, biomarkers are lacking and treatment remains expectant and supportive. This study aimed to understand the pathophysiological mechanisms driving SIRS induced by CPB and identify novel therapeutic targets that might reduce systemic inflammation and improve patient outcomes. Methods: Twenty-one patients undergoing cardiac surgery and CPB were recruited, and blood was sampled before, during and after surgery. SIRS was defined using the American College of Chest Physicians/Society of Critical Care Medicine criteria. We performed immune cell profiling and whole blood transcriptomics and measured individual mediators in plasma/serum to characterise SIRS induced by CPB. Results: Nineteen patients fulfilled criteria for SIRS, with a mean duration of 2.7 days. Neutrophil numbers rose rapidly with CPB and remained elevated for at least 48 h afterwards. Transcriptional signatures associated with neutrophil activation and degranulation were enriched during CPB. We identified a network of cytokines governing these transcriptional changes, including granulocyte colony-stimulating factor (G-CSF), a regulator of neutrophil production and function. Conclusions: We identified neutrophils and G-CSF as major regulators of CPB-induced systemic inflammation. Short-term targeting of G-CSF could provide a novel therapeutic strategy to limit neutrophil-mediated inflammation and tissue damage in SIRS induced by CPB.

2.
iScience ; 26(7): 107173, 2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37456846

RESUMO

Systemic lupus erythematosus (SLE) is an autoimmune disease with significant morbidity and mortality. Type I interferon (IFN) drives SLE pathology and plasmacytoid dendritic cells (pDCs) are potent producers of IFN; however, the specific effects of pDC depletion have not been demonstrated. We show CD123 was highly expressed on pDCs and the anti-CD123 antibody CSL362 potently depleted pDCs in vitro. CSL362 pre-treatment abrogated the induction of IFNα and IFN-induced gene transcription following stimulation with SLE patient-derived serum or immune complexes. RNA transcripts induced in pDCs by ex vivo stimulation with TLR ligands were reflected in gene expression profiles of SLE blood, and correlated with disease severity. TLR ligand-induced protein production by SLE patient peripheral mononuclear cells was abrogated by CSL362 pre-treatment including proteins over expressed in SLE patient serum. These findings implicate pDCs as key drivers in the cellular activation and production of soluble factors seen in SLE.

3.
Immunotherapy ; 9(3): 249-259, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28183192

RESUMO

AIM: Pilot clinical trial of NY-ESO-1 (ESO) protein in ISCOMATRIX™ adjuvant pulsed onto peripheral blood dendritic cells (PBDC), to ascertain feasibility, evaluate toxicity and assess induction of ESO-specific immune responses. PATIENTS & METHODS: Eligible participants had resected cancers expressing ESO or LAGE-1 and were at high risk of relapse. PBDC were produced using CliniMACS®plus, with initial depletion of CD1c+ B cells followed by positive selection of CD1c+ PBDC. Patients received three intradermal vaccinations of ESO/IMX-pulsed PBDC at 4-week intervals. RESULTS: The process was feasible and safe. No vaccine-induced immune responses were detected. Assays of immunomodulatory cells did not correlate with outcomes. One patient had a long lasting complete remission. CONCLUSION: This method was feasible and safe but was minimally immunogenic.


Assuntos
Células Sanguíneas/fisiologia , Vacinas Anticâncer/imunologia , Carcinoma Basocelular/terapia , Células Dendríticas/fisiologia , Imunoterapia/métodos , Neoplasias Cutâneas/terapia , Linfócitos T/imunologia , Idoso , Apresentação de Antígeno , Antígenos CD1/metabolismo , Antígenos de Neoplasias/metabolismo , Células Sanguíneas/transplante , Carcinoma Basocelular/imunologia , Carcinoma Basocelular/patologia , Células Cultivadas , Colesterol/metabolismo , Células Dendríticas/transplante , Combinação de Medicamentos , Feminino , Seguimentos , Glicoproteínas/metabolismo , Humanos , Imunidade Humoral , Ativação Linfocitária , Masculino , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Metástase Neoplásica , Estadiamento de Neoplasias , Fosfolipídeos/metabolismo , Projetos Piloto , Saponinas/metabolismo , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Resultado do Tratamento
4.
JCI Insight ; 1(6): e86131, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-27699260

RESUMO

To date, the major target of biologic therapeutics in systemic lupus erythematosus (SLE) has been the B cell, which produces pathogenic autoantibodies. Recently, targeting type I IFN, which is elaborated by plasmacytoid dendritic cells (pDCs) in response to endosomal TLR7 and TLR9 stimulation by SLE immune complexes, has shown promising results. pDCs express high levels of the IL-3Rα chain (CD123), suggesting an alternative potential targeting strategy. We have developed an anti-CD123 monoclonal antibody, CSL362, and show here that it affects key cell types and cytokines that contribute to SLE. CSL362 potently depletes pDCs via antibody-dependent cell-mediated cytotoxicity, markedly reducing TLR7, TLR9, and SLE serum-induced IFN-α production and IFN-α-upregulated gene expression. The antibody also inhibits TLR7- and TLR9-induced plasmablast expansion by reducing IFN-α and IL-6 production. These effects are more pronounced than with IFN-α blockade alone, possibly because pDC depletion reduces production of other IFN subtypes, such as type III, as well as non-IFN proinflammatory cytokines, such as IL-6. In addition, CSL362 depletes basophils and inhibits IL-3 signaling. These effects were confirmed in cells derived from a heterogeneous population of SLE donors, various IFN-dependent autoimmune diseases, and healthy controls. We also demonstrate in vivo activity of CSL362 following its s.c. administration to cynomolgus monkeys. This spectrum of effects provides a preclinical rationale for the therapeutic evaluation of CSL362 in SLE.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Células Dendríticas/imunologia , Subunidade alfa de Receptor de Interleucina-3/imunologia , Lúpus Eritematoso Sistêmico/terapia , Anticorpos Monoclonais/imunologia , Complexo Antígeno-Anticorpo , Células Cultivadas , Humanos , Interferon-alfa/sangue , Interleucina-6/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Receptor 7 Toll-Like/imunologia , Receptor Toll-Like 9/imunologia
5.
Cancer Immun ; 5: 5, 2005 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-15755075

RESUMO

Immune responses to cancer vaccines are commonly tested by measuring cutaneous reactions to intradermal (i.d.) antigen. When well-characterized peptide epitopes are injected i.d., infiltrates of CD4+ and CD8+ T lymphocytes are frequently seen. In this study, we have further characterized T cells derived from vaccine-infiltrating lymphocyte (VIL) responses. We found that the infiltrates capable of producing IFN-gamma and cytolytic activity could recognize vaccine peptide, as well as antigen-positive melanoma cells. We studied antigen-specific T cell responses from VILs and peripheral blood in 10 patients who participated in a clinical trial. All patients received systemic Flt3 ligand (20 microg/kg/d) and i.d. peptides: Three NY-ESO-1 peptides, SLLMWITQCFL (157-167), SLLMWITQC (157-165), QLSLLMWIT (155-163); tyrosinase internal peptide YMDGTMSQV (368-376); Melan-A/MART-1 analogue peptide ELAGIGILTV (26-35, E27L substitution); and influenza matrix peptide GILGFVFTL (58-66). In 54 paired VIL and peripheral blood analyses, a good correlation was found between responses in skin and in blood. These cells could be rapidly expanded in a short-term assay and thus appear to be memory T cells. The demonstrated presence of antigen-specific T cells at vaccination sites validates this method of assessing the immune response to i.d. vaccines.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Hipersensibilidade Tardia/imunologia , Linfócitos do Interstício Tumoral/imunologia , Células Sanguíneas/imunologia , Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Testes Imunológicos de Citotoxicidade , Humanos , Injeções Intradérmicas , Interferon gama/biossíntese , Melanoma/imunologia , Pele/imunologia , Neoplasias Cutâneas/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
6.
Clin Cancer Res ; 10(8): 2879-90, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15102697

RESUMO

NY-ESO-1 is a 180 amino-acid human tumor antigen expressed by many different tumor types and belongs to the family of "cancer-testis" antigens. In humans, NY-ESO-1 is one of the most immunogenic tumor antigens and NY-ESO-1 peptides have been shown to induce NY-ESO-1-specific CD8(+) CTLs capable of altering the natural course of NY-ESO-1-expressing tumors in cancer patients. Here we describe the preclinical immunogenicity and efficacy of NY-ESO-1 protein formulated with the ISCOMATRIX adjuvant (NY-ESO-1 vaccine). In vitro, the NY-ESO-1 vaccine was readily taken up by human monocyte-derived dendritic cells, and on maturation, these human monocyte-derived dendritic cells efficiently cross-presented HLA-A2-restricted epitopes to NY-ESO-1-specific CD8(+) T cells. In addition, epitopes of NY-ESO-1 protein were also presented on MHC class II molecules to NY-ESO-1-specific CD4(+) T cells. The NY-ESO-1 vaccine induced strong NY-ESO-1-specific IFN-gamma and IgG2a responses in C57BL/6 mice. Furthermore, the NY-ESO-1 vaccine induced NY-ESO-1-specific CD8(+) CTLs in HLA-A2 transgenic mice that were capable of lysing human HLA-A2(+) NY-ESO-1(+) tumor cells. Finally, C57BL/6 mice, immunized with the NY-ESO-1 vaccine, were protected against challenge with a B16 melanoma cell line expressing NY-ESO-1. These data illustrate that the NY-ESO-1 vaccine represents a potent therapeutic anticancer vaccine.


Assuntos
Adjuvantes Imunológicos , Antígenos de Neoplasias/química , Antineoplásicos/farmacologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer , Imunidade Celular , Proteínas de Membrana/química , Animais , Antígenos CD4/biossíntese , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Epitopos/química , Escherichia coli/metabolismo , Antígeno HLA-A2/química , Imunoglobulina G/química , Imuno-Histoquímica , Melanoma Experimental , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/metabolismo , Peptídeos/química , Fenótipo , Plasmídeos/metabolismo , RNA Mensageiro/metabolismo , Proteínas Recombinantes/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção
7.
J Immunol ; 176(10): 5908-17, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16670298

RESUMO

Immunodominance has been well-demonstrated in many antiviral and antibacterial systems, but much less so in the setting of immune responses against cancer. Tumor Ag-specific CD8+ T cells keep cancer cells in check via immunosurveillance and shape tumor development through immunoediting. Because most tumor Ags are self Ags, the breadth and depth of antitumor immune responses have not been well-appreciated. To design and develop antitumor vaccines, it is important to understand the immunodominance hierarchy and its underlying mechanisms, and to identify the most immunodominant tumor Ag-specific T cells. We have comprehensively analyzed spontaneous cellular immune responses of one individual and show that multiple tumor Ags are targeted by the patient's immune system, especially the "cancer-testis" tumor Ag NY-ESO-1. The pattern of anti-NY-ESO-1 T cell responses in this patient closely resembles the classical broad yet hierarchical antiviral immunity and was confirmed in a second subject.


Assuntos
Antígenos de Neoplasias/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos Imunodominantes/imunologia , Proteínas de Membrana/fisiologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Células CHO , Linhagem Celular Transformada , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Humanos , Imunidade Inata , Epitopos Imunodominantes/metabolismo , Masculino , Pessoa de Meia-Idade
8.
J Immunother ; 29(5): 499-511, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16971806

RESUMO

Flt3 ligand mobilizes dendritic cells (DCs) into blood, allowing generation in vivo of large numbers of DCs for immunotherapy. These immature DCs can be rapidly activated by soluble CD40 ligand (CD40L). We developed a novel overnight method using these cytokines to produce DCs for cancer immunotherapy. Flt3 ligand-mobilized DCs (FLDCs) were isolated, activated with CD40L, loaded with antigenic peptides from influenza matrix protein, hepatitis B core antigen, NY-ESO-1, MAGE-A4, and MAGE-A10, and injected into patients with resected melanoma. Three injections were given at 4-week intervals. Study end points included antigen-specific immune responses (skin reactions to peptides alone or peptide-pulsed FLDCs; circulating T-cell responses), safety, and toxicity. No patient had a measurable tumor. Six patients were entered. FLDCs were obtained, enriched, and cultured under Good Manufacturing Practice grade conditions. Overnight culture with soluble CD40L caused marked up-regulation of activation markers (CD83 and HLA-DR). These FLDCs were functional and able to stimulate antigen-specific T cells in vitro. No significant adverse events were attributable to FLDCs. Peptide-pulsed FLDCs caused strong local skin reactions up to 60 mm diameter with intense perivascular infiltration of T cells, exceeding those seen in our previous peptide-based protocols. Antigen-specific blood T-cell responses were induced, including responses to an antigen for which the patients were naive (hepatitis B core antigen) and MAGE-A10. MAGE-A10-specific T cells with a skewed T-cell receptor repertoire were detected in 1 patient in blood ex vivo and from tumor biopsies. Vaccination with FLDCs pulsed with peptides is safe and primes immune responses to cancer antigens.


Assuntos
Ligante de CD40/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Melanoma/terapia , Proteínas de Membrana/imunologia , Neoplasias Cutâneas/terapia , Adulto , Apresentação de Antígeno , Antígenos CD8/imunologia , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Células Cultivadas , Citotoxicidade Imunológica , Feminino , Humanos , Imunoterapia Adotiva , Leucaférese , Ativação Linfocitária , Masculino , Melanoma/imunologia , Pessoa de Meia-Idade , Peptídeos/imunologia , Peptídeos/uso terapêutico , Projetos Piloto , Receptores de Antígenos de Linfócitos T/metabolismo , Neoplasias Cutâneas/imunologia
9.
Eur J Immunol ; 34(10): 2919-29, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15368308

RESUMO

NY-ESO-1 is a germ cell antigen aberrantly expressed by different tumor types that elicits strong humoral and cellular immune responses, representing one of the most promising candidates for vaccination of cancer patients. A detailed analysis of CD8(+) T cells generated in vaccine trials using NY-ESO-1-derived peptides (157-165 and 157-167) revealed that the dominant immune response was directed against a cryptic epitope (159-167) diverting the immune response from tumor recognition. Only CTL reactivity to the NY-ESO-1(157-165) peptide appeared to be capable of lysing NY-ESO-1/HLA-A0201-expressing tumor cells. To study the process of NY-ESO-1 peptide presentation by tumor cells in more detail we generated a high-affinity (K(D)=60 nM) antibody fragment that specifically recognizes the NY-ESO-1(157-165) peptide/HLA-A0201 complex. Peptide variants such as the NY-ESO-1(157-167) peptide or the cryptic NY-ESO-1(159-167) peptide were not recognized. The antibody fragment blocked in a dose-dependent fashion the recognition of NY-ESO-1/HLA-A2-positive tumor cells by NY-ESO-1(157-165) peptide-specific CD8(+) T cells. This antibody fragment is a novel reagent that binds with TCR-like specificity to the NY-ESO-1(157-165)/HLA-A2 complex thus distinguishing between CTL responses against immunological meaningful or cryptic NY-ESO-1-derived peptides. It may therefore become a useful monitoring tool for the development of NY-ESO-1-based cancer vaccines.


Assuntos
Especificidade de Anticorpos/imunologia , Antígenos de Neoplasias/imunologia , Fragmentos Fab das Imunoglobulinas/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Proteínas de Membrana/imunologia , Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Sequência de Aminoácidos , Apresentação de Antígeno/imunologia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Epitopos Imunodominantes/imunologia , Conformação Proteica , Transfecção
10.
Blood ; 102(5): 1753-63, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12738673

RESUMO

Dendritic cells (DCs) are a family of leukocytes that initiate T- and B-cell immunity against pathogens. Migration of antigen-loaded DCs from sites of infection into draining lymphoid tissues is fundamental to the priming of T-cell immune responses. In humans, the major peripheral blood DC (PBDC) types, CD1c+ DCs and interleukin 3 receptor-positive (IL-3R+) plasmacytoid DCs, are significantly expanded in vivo with the use of Flt3 ligand (FL). DC-like cells can also be generated from monocyte precursors (MoDCs). A detailed comparison of the functional potential of these types of DCs (in an autologous setting) has yet to be reported. Here, we compared the functional capacity of FL-expanded CD1c+ PBDCs with autologous MoDCs in response to 3 different classes of stimuli: (1) proinflammatory mediators, (2) soluble CD40 ligand trimer (CD40L), and (3) intact bacteria (Escherichia coli). Significant differences in functional capacities were found with respect to changes in phenotype, migratory capacity, cytokine secretion, and T-cell stimulation. MoDCs required specific stimuli for the expression of functions. They responded vigorously to CD40L or E coli, expressing cytokines known to regulate interferon-gamma (IFN-gamma) in T cells (IL-12p70, IL-18, and IL-23), but required prostaglandin E2 (PGE2) during stimulation to migrate to chemokines. In contrast, PBDCs matured in response to minimal stimulation, rapidly acquired migratory function in the absence of PGE2-containing stimuli, and were low cytokine producers. Interestingly, both types of DCs were equivalent with respect to stimulation of allogeneic T-cell proliferation and presentation of peptides to cytotoxic T lymphocyte (CTL) lines. These distinct differences are of particular importance when considering the choice of DC types for clinical applications.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/imunologia , Proteínas de Membrana/farmacologia , Monócitos/citologia , Antígenos CD1/metabolismo , Ligante de CD40/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Divisão Celular/imunologia , Movimento Celular/imunologia , Células Cultivadas , Citocinas/biossíntese , Células Dendríticas/metabolismo , Escherichia coli , Glicoproteínas/metabolismo , Humanos , Imunofenotipagem , Técnicas In Vitro , Mediadores da Inflamação/farmacologia , Ativação Linfocitária/imunologia , Melanoma/imunologia , Peptídeos/farmacologia , Estimulação Química , Linfócitos T/citologia , Linfócitos T/imunologia
11.
Proc Natl Acad Sci U S A ; 101(25): 9363-8, 2004 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-15197261

RESUMO

There is increasing evidence showing the involvement of CD4(+) T cells in initiating and maintaining antitumor immune responses. NY-ESO-1 is expressed by various tumors but not normal tissues except testis. We conducted a cancer clinical trial by using full-length NY-ESO-1 protein formulated with ISCOMATRIX adjuvant and injected into patients intramuscularly. Autologous dendritic cells pulsed with NY-ESO-1 ISCOMATRIX in combination with overlapping synthetic peptides were used to identify immunodominant T cells from a vaccinated patient. We show here the identification and characterization of two novel CD4(+) T cell epitopes. T cells specific to these epitopes not only recognized autologous dendritic cells loaded with NY-ESO-1 but also NY-ESO-1-expressing tumor cell lines treated with IFN-gamma. One of the two responses identified was greater than the previously identified immunodominant HLA-DP4-restricted response and correlated with NY-ESO-1-specific CD8(+) T cell induction after vaccination. This T cell response was vaccinated in most patients who expressed HLA-DR2. This study has systematically surveyed patients vaccinated with full-length tumor antigen for a vaccinated CD4 helper T cell response.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Proteínas de Membrana/imunologia , Vacinas Sintéticas/imunologia , Sequência de Aminoácidos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Epitopos/química , Epitopos/imunologia , Humanos , Ativação Linfocitária , Complexo Principal de Histocompatibilidade/imunologia , Masculino , Dados de Sequência Molecular , Testículo/imunologia
12.
Proc Natl Acad Sci U S A ; 101(29): 10697-702, 2004 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-15252201

RESUMO

NY-ESO-1 is a "cancer-testis" antigen expressed in many cancers. ISCOMATRIX is a saponin-based adjuvant that induces antibody and T cell responses. We performed a placebo-controlled clinical trial evaluating the safety and immunogenicity of recombinant NY-ESO-1 protein with ISCOMATRIX adjuvant. Forty-six evaluable patients with resected NY-ESO-1-positive tumors received three doses of vaccine intramuscularly at monthly intervals. The vaccine was well tolerated. We observed high-titer antibody responses, strong delayed-type hypersensitivity reactions, and circulating CD8(+) and CD4(+) T cells specific for a broad range of NY-ESO-1 epitopes, including known and previously unknown epitopes. In an unplanned analysis, vaccinated patients appeared to have superior clinical outcomes to those treated with placebo or protein alone. The vaccine is safe and highly potent immunologically.


Assuntos
Adjuvantes Imunológicos , Anticorpos Antineoplásicos/imunologia , Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer , Proteínas de Membrana/imunologia , Proteínas Recombinantes/imunologia , Saponinas/imunologia , Antígenos de Neoplasias/genética , Relação Dose-Resposta a Droga , Método Duplo-Cego , Epitopos , Humanos , Masculino , Melanoma/imunologia , Melanoma/patologia , Proteínas de Membrana/genética , Placebos , Proteínas Recombinantes/genética , Testículo/patologia , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA