Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Leukemia ; 33(8): 1923-1933, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30728457

RESUMO

The aim of this randomized phase-II study was to evaluate the effect of substituting cytarabine by azacitidine in intensive induction therapy of patients with acute myeloid leukemia (AML). Patients were randomized to four induction schedules for two cycles: STANDARD (idarubicin, cytarabine, etoposide); and azacitidine given prior (PRIOR), concurrently (CONCURRENT), or after (AFTER) therapy with idarubicin and etoposide. Consolidation therapy consisted of allogeneic hematopoietic-cell transplantation or three courses of high-dose cytarabine followed by 2-year maintenance therapy with azacitidine in the azacitidine-arms. AML with CBFB-MYH11, RUNX1-RUNX1T1, mutated NPM1, and FLT3-ITD were excluded and accrued to genotype-specific trials. The primary end point was response to induction therapy. The statistical design was based on an optimal two-stage design applied for each arm separately. During the first stage, 104 patients (median age 62.6, range 18-82 years) were randomized; the study arms PRIOR and CONCURRENT were terminated early due to inefficacy. After randomization of 268 patients, all azacitidine-containing arms showed inferior response rates compared to STANDARD. Event-free and overall survival were significantly inferior in the azacitidine-containing arms compared to the standard arm (p < 0.001 and p = 0.03, respectively). The data from this trial do not support the substitution of cytarabine by azacitidine in intensive induction therapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Azacitidina/administração & dosagem , Etoposídeo/administração & dosagem , Feminino , Humanos , Idarubicina/administração & dosagem , Quimioterapia de Indução , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidade , Masculino , Pessoa de Meia-Idade , Nucleofosmina , Estudos Prospectivos , Adulto Jovem
2.
Ann Hematol ; 96(12): 1993-2003, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29090343

RESUMO

We describe genetic and clinical characteristics of acute myeloid leukemia (AML) patients according to age from an academic population-based registry. Adult patients with newly diagnosed AML at 63 centers in Germany and Austria were followed within the AMLSG BiO registry (NCT01252485). Between January 1, 2012, and December 31, 2014, data of 3525 patients with AML (45% women) were collected. The median age was 65 years (range 18-94). The comparison of age-specific AML incidence rates with epidemiological cancer registries revealed excellent coverage in patients < 70 years old and good coverage up to the age of 80. The distribution according to the European LeukemiaNet (ELN) risk categorization from 2010 was 20% favorable, 31% intermediate-1, 28% intermediate-2, and 21% adverse. With increasing age, the relative but not the absolute prevalence of patients with ELN favorable and intermediate-1 risk (p < 0.001), with activating FLT3 mutations (p < 0.001), with ECOG performance status < 2 (p < 0.001), and with HCT-CI comorbidity index < 3 (p < 0.001) decreased. Regarding treatment, obesity and favorable risk were associated with an intensive treatment, whereas adverse risk, higher age, and comorbidity index > 0 were associated with non-intensive treatment or best supportive care. The AMLSG BiO registry provides reliable population-based distributions of genetic, clinical, and treatment characteristics according to age.


Assuntos
Leucemia Mieloide Aguda , Mutação , Sistema de Registros , Tirosina Quinase 3 Semelhante a fms , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Áustria , Feminino , Alemanha , Humanos , Leucemia Mieloide Aguda/epidemiologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/terapia , Masculino , Pessoa de Meia-Idade , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
4.
Leukemia ; 31(11): 2347-2354, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28322237

RESUMO

Therapy-related acute promyelocytic leukemia (t-APL) is relatively rare, with limited data on outcome after treatment with arsenic trioxide (ATO) compared to standard intensive chemotherapy (CTX). We evaluated 103 adult t-APL patients undergoing treatment with all-trans retinoic acid (ATRA) alone (n=7) or in combination with ATO (n=24), CTX (n=53), or both (n=19). Complete remissions were achieved after induction therapy in 57% with ATRA, 100% with ATO/ATRA, 78% with CTX/ATRA, and 95% with CTX/ATO/ATRA. Early death rates were 43% for ATRA, 0% for ATO/ATRA, 12% for CTX/ATRA and 5% for CTX/ATO/ATRA. Three patients relapsed, two developed therapy-related acute myeloid leukemia and 13 died in remission including seven patients with recurrence of the prior malignancy. Median follow-up for survival was 3.7 years. None of the patients treated with ATRA alone survived beyond one year. Event-free survival was significantly higher after ATO-based therapy (95%, 95% CI, 82-99%) as compared to CTX/ATRA (78%, 95% CI, 64-87%; P=0.042), if deaths due to recurrence of the prior malignancy were censored. The estimated 2-year overall survival in intensively treated patients was 88% (95% CI, 80-93%) without difference according to treatment (P=0.47). ATO when added to ATRA or CTX/ATRA is feasible and leads to better outcomes as compared to CTX/ATRA in t-APL.


Assuntos
Arsenicais/uso terapêutico , Leucemia Promielocítica Aguda/tratamento farmacológico , Segunda Neoplasia Primária/tratamento farmacológico , Óxidos/uso terapêutico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Trióxido de Arsênio , Feminino , Humanos , Leucemia Promielocítica Aguda/etiologia , Leucemia Promielocítica Aguda/genética , Masculino , Pessoa de Meia-Idade , Segunda Neoplasia Primária/etiologia , Segunda Neoplasia Primária/genética , Indução de Remissão , Análise de Sobrevida , Resultado do Tratamento , Adulto Jovem
5.
Cancer Gene Ther ; 23(6): 162-7, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27056671

RESUMO

Chemokines are key regulators of both innate and adaptive immune responses. CCL4 (macrophage inflammatory protein-1ß, MIP-1ß) is a CC chemokine that has a broad spectrum of target cells including immature dendritic cells, which express the cognate receptor CCR5. We asked whether a plasmid encoding CCL4 is able to improve tumor protection and immune responses in a Her2/neu+ mouse tumor model. Balb/c mice were immunized twice intramuscularly with plasmid DNA on days 1 and 15. On day 25, a tumor challenge was performed with 2 × 10(5) syngeneic Her2/neu+ D2F2/E2 tumor cells. Different groups of mice were vaccinated with pDNA(Her2/neu) plus pDNA(CCL4), pDNA(Her2/neu), pDNA(CCL4) or mock vector alone. Our results show that CCL4 is able to (i) improve tumor protection and (ii) augment a TH1-polarized immune response against Her2/neu. Although Her2/neu-specific humoral and T-cell immune responses were comparable with that induced in previous studies using CCL19 or CCL21 as adjuvants, tumor protection conferred by CCL4 was inferior. Whether this is due to a different spectrum of (innate) immune cells, remains to be clarified. However, combination of CCL19/21 with CCL4 might be a reasonable approach in the future, particularly for DNA vaccination in Her2/neu+ breast cancer in the situation of minimal residual disease.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer/imunologia , Quimiocina CCL4 , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Receptor ErbB-2/genética , Vacinas de DNA/imunologia , Animais , Vacinas Anticâncer/genética , Linhagem Celular , Quimiocina CCL4/genética , Modelos Animais de Doenças , Feminino , Expressão Gênica , Ordem dos Genes , Humanos , Imunização , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Plasmídeos/genética , Carga Tumoral , Vacinas de DNA/genética
6.
Cancer Gene Ther ; 19(12): 880-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23099886

RESUMO

The aim of this study was to evaluate the efficacy of the chemokine CCL19 (ELC) as an adjuvant for intradermal gene gun delivery of Her2/neu DNA and to investigate the role of B cells in CCL19-mediated enhancement of immune responses. Balb/c mice were immunized intramuscularly (i.m.) on days 1 and 15 with plasmid DNA (pDNA) (100 µg DNA) or intradermally (i.d.) by gene gun delivery (1-2 µg DNA). Administration of pDNA encoding Her2/neu (pDNA(Her2/neu) was compared with pDNA(Her2/neu) plus pDNA(CCL19), pDNA(CCL19), mock vector or uncoated gold particles/phosphate-buffered saline (PBS). Tumor challenge was performed subcutaneously on day 25 with syngeneic Her2/neu(+) tumor cells (D2F2/E2). Intradermal immunization by gene gun led to an enhancement of tumor protection by the DNA vaccine as compared with i.m. immunization. The protective effect of the vaccine was further enhanced by coadministration of pDNA(CCL19) both after i.m. and i.d. immunization. Tumor protection was associated with Her2/neu-specific T cell and humoral immune responses. Experiments in B-cell-deficient µMT mice showed that B cells are crucial for CCL19-mediated enhancement of tumor rejection, most likely as antigen-presenting B cells. DNA vaccines against Her2/neu may play a future role in the treatment of Her2/neu-positive breast cancer patients in a clinical situation of minimal residual disease.


Assuntos
Linfócitos B/imunologia , Biolística/métodos , Vacinas Anticâncer/administração & dosagem , Quimiocina CCL19/genética , Quimiocina CCL19/imunologia , Neoplasias Mamárias Experimentais/terapia , Receptor ErbB-2/imunologia , Vacinas de DNA/administração & dosagem , Animais , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Quimiocina CCL19/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Injeções Intradérmicas , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Receptor ErbB-2/genética , Linfócitos T/imunologia , Vacinas de DNA/genética , Vacinas de DNA/imunologia
7.
Cancer Gene Ther ; 19(1): 69-76, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21997231

RESUMO

Secondary lymphoid-tissue chemokine (SLC/CCL21) is a CC chemokine that is constitutively expressed in various lymphoid tissues and binds to chemokine receptor CCR7 on mature dendritic cells (DCs) and distinct T-and B-cell sub-populations. In vivo, CCL21 regulates the encounters between DC and T cells and thus is a key regulator of adaptive immune responses. We asked whether CCL21 is able to augment immunogenicity of a DNA-based vaccine against Her2/neu in a Balb/c mouse model with syngeneic Her2/neu+ tumor cells (D2F2/E2). Mice were vaccinated intramuscularly with plasmid DNA (pDNA) on day 1 and boosted on day 15; tumor challenge was performed subcutaneously on day 25. Coexpression of CCL21 and Her-2/neu resulted in induction of a TH1-polarized immune response and substantial improvement of the protective effect of the DNA vaccine. Coexpression of tumor antigen pDNA(Her2/neu) with both pDNA(GM-CSF) and pDNA(CCL21) as adjuvants led to further improvement of protection by the vaccine (70% tumor-free mice on day 35 vs 40% with either adjuvant alone vs 5-10% with tumor antigen alone). Our results show that CCL21 is a potent adjuvant for DNA vaccination, particularly in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF). Clinical use of a pDNA(Her2/neu/CCL21/GM-CSF) vaccine might be particularly promising in minimal residual Her2/neu+ breast cancer.


Assuntos
Vacinas Anticâncer/imunologia , Quimiocina CCL21/imunologia , Neoplasias Mamárias Experimentais/prevenção & controle , Receptor ErbB-2/imunologia , Vacinas de DNA/imunologia , Animais , Vacinas Anticâncer/genética , Vacinas Anticâncer/farmacologia , Linhagem Celular Tumoral , Quimiocina CCL21/genética , Modelos Animais de Doenças , Feminino , Humanos , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Receptor ErbB-2/biossíntese , Receptor ErbB-2/genética , Vacinas de DNA/genética , Vacinas de DNA/farmacologia
8.
Gene Ther ; 18(4): 354-63, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21068778

RESUMO

Despite novel targeted agents, prognosis of metastatic renal cell cancer (RCC) remains poor, and experimental therapeutic strategies are warranted. Transfection of tumor cells with co-stimulatory molecules and/or cytokines is able to increase immunogenicity. Therefore, in our clinical study, 10 human leukocyte antigen (HLA)-A(*)0201(+) patients with histologically-confirmed progressive metastatic clear cell RCC were immunized repetitively over 22 weeks with 2.5-40 × 10(6) interleukin (IL)-7/CD80 cotransfected allogeneic HLA-A(*)0201(+) tumor cells (RCC26/IL-7/CD80). Endpoints of the study were feasibility, safety, immunological and clinical responses. Vaccination was feasible and safe. In all, 50% of the patients showed stable disease throughout the study; the median time to progression was 18 weeks. However, vaccination with allogeneic RCC26/IL-7/CD80 tumor cells was not able to induce TH1-polarized immune responses. A TH2 cytokine profile with increasing amounts of antigen-specific IL-10 secretion was observed in most of the responding patients. Interferon-γ secretion by patient lymphocytes upon antigen-specific and non-specific stimulation was substantially impaired, both before and during vaccination, as compared with healthy controls. This is possibly due to profound tumor-induced immunosuppression, which may prevent induction of antitumor immune responses by the gene-modified vaccine. Vaccination in minimal residual disease with concurrent depletion of regulatory cells might be one strategy to overcome this limitation.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma de Células Renais/terapia , Interleucina-7/imunologia , Neoplasias Renais/terapia , Adulto , Idoso , Antígeno B7-1/metabolismo , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Feminino , Antígenos HLA/análise , Humanos , Masculino , Pessoa de Meia-Idade , Linfócitos T/imunologia , Transfecção
9.
J Anat ; 212(2): 114-24, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18194204

RESUMO

The reciprocal influences of thymic lymphocyte and nonlymphocyte populations, i.e. thymic cross-talk, are necessary for the proper maturation of thymocytes and the development/maintenance of thymic stromal microenvironments. Although the molecular influences exerted by thymic stromal cells on maturing thymocytes have been extensively studied, the identity of signalling molecules used by thymocytes to influence the thymic stromal cells is still largely unknown. Our study provides the first ultrastructural evidence that the functional lymphotoxin-beta receptor (LTbetaR) signalling pathway is engaged in the cross-talk between thymocytes and the thymic stromal cell population. We show that LTbetaR signalling is of the utmost significance for the preservation of the subcellular integrity of all thymic epithelial cells. In the absence of LTbetaR there is (1) hypertrophy and activation of cortical thymic epithelial cells, (2) the complete loss of fully differentiated medullary thymic epithelial cells, and (3) the inhibited differentiation of remaining medullary thymic epithelial cells with the appearance of prominent intercellular cysts in the thymic medulla.


Assuntos
Células Epiteliais/diagnóstico por imagem , Receptor beta de Linfotoxina/deficiência , Microscopia Eletrônica/métodos , Timo/ultraestrutura , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ultrassonografia
10.
Cancer Gene Ther ; 14(6): 523-32, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17384577

RESUMO

Coexpression of tumor antigens together with immunomodulatory molecules is a strategy in DNA vaccination aiming at an amplification of the antitumor immune response. Epstein-Barr virus-induced-molecule-1-ligand-chemokine (ELC/CCL19) is a CC chemokine that binds to the chemokine receptor CCR7. CCR7 is expressed on mature dendritic cells (DC) and distinct T- and B-cell subpopulations. CCL19 (ELC) is mainly expressed in secondary lymphoid organs and plays a central role in regulating the encounters between DC and T cells. We asked whether CCL19 is able to augment immunogenicity of a DNA vaccine in a C57BL/6 mouse model with syngeneic MCA205 (beta-gal) tumor cells. Mice were vaccinated twice intramuscularly on days 1 and 15 and tumor challenge was performed subcutaneously on day 25. Coadministration of plasmid DNA (pDNA) (beta-gal) plus pDNA (CCL19) was compared with pDNA (beta-gal), pDNA (CCL19), mock vector and phosphate-buffered saline (PBS) alone. Coexpression of CCL19 resulted in enhancement of a Th1-polarized immune response with substantial improvement of the protective effect of the DNA vaccine. Immunohistochemical staining revealed an increased CD8+ T-cell infiltration in the tumor tissue of mice that had been immunized with pDNA (beta-gal) plus pDNA (CCL19). We conclude that CCL19 is an attractive adjuvant for DNA vaccination able to augment antitumor immunity and that this effect is partially caused by enhanced CD8+ T-cell recruitment.


Assuntos
Antineoplásicos/imunologia , Quimiocinas CC/imunologia , Células Dendríticas/efeitos dos fármacos , Neoplasias/terapia , Linfócitos T/efeitos dos fármacos , Vacinas de DNA/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Quimiocina CCL19 , Quimiocinas CC/administração & dosagem , Células Dendríticas/imunologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Linfócitos T/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Vacinação , Vacinas de DNA/administração & dosagem
11.
Clin Exp Immunol ; 126(3): 421-5, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11737056

RESUMO

Although many studies focus on the increase of immunocompetent cells within the middle ear mucosa during acute otitis media it is poorly understood how this increase is mediated. The differentiation between two possible causes, i.e. immigration and local proliferation, would help to better understand the pathophysiology of this disease. Therefore, the number of proliferating macrophages, dendritic cells, natural killer cells and T and B lymphocytes was studied during acute otitis media in the rat middle ear mucosa (ME mucosa) and Eustachian tube mucosa (ET mucosa) by labelling proliferating leucocytes with the DNA precursor bromodeoxyuridine (BrdU). By removing the middle ear and Eustachian tube 24 h after BrdU injection, the contribution of immigrated newly formed cells was estimated. At this timepoint, many leucocytes in the ME and ET mucosa had incorporated BrdU (between 15 and 25% within the subsets). By analysing these tissues one hour after BrdU injection, the local proliferation rate was determined (between 2 and 9% within the subsets). Thus, the inflamed ME and ET mucosa are the destination of immunocompetent cells and, as our data show, the inflamed microenvironment supports local proliferation of immunocompetent cells.


Assuntos
Orelha Média/imunologia , Orelha Média/patologia , Otite Média Supurativa/imunologia , Otite Média Supurativa/patologia , Doença Aguda , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Bromodesoxiuridina/metabolismo , Divisão Celular , DNA/biossíntese , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Orelha Média/metabolismo , Tuba Auditiva/imunologia , Tuba Auditiva/metabolismo , Tuba Auditiva/patologia , Feminino , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Mucosa/imunologia , Mucosa/metabolismo , Mucosa/patologia , Otite Média Supurativa/metabolismo , Ratos , Ratos Endogâmicos Lew , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia
12.
Blood ; 98(10): 3035-41, 2001 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11698288

RESUMO

Splenectomy increases the number of B cells in the blood of humans and animals. It is unknown whether this is due to changes in migration, proliferation, or both. The numbers of naïve (IgD(+)IgM(+)), memory (IgD(-)IgM(high)), newly formed (IgM(high)CD90(high)), early recirculating follicular (IgM(low)CD90(high)), recirculating follicular (IgM(low)CD90(-)), and marginal zone (IgM(high)CD90(-)) phenotype B cells were determined in control and splenectomized rats by flow cytometry. All subsets increased significantly in the blood after splenectomy. Because surface molecules are involved in the regulation of migration and proliferation, their expression (lymphocyte function-associated antigen 1 [LFA-1], intercellular adhesion molecule 1 (ICAM-1), L-selectin, alpha4-integrins, CD44, major histocompatability complex class II, interleukin 2 receptor-alpha chain) was determined on B- and T-cell subsets of both groups. B cells, but not T cells, showed a significantly reduced LFA-1 and ICAM-1 expression in blood and lymph nodes, whereas the expression of the other surface molecules analyzed remained unchanged. The down-regulation of these molecules did not influence the adherence of B cells to high endothelial venules in vitro. In vivo, however, ICAM-1(low)-expressing B cells migrated significantly faster through lymph nodes (ICAM-1(low) 41 +/- 5 hours versus ICAM-1(high) 58 +/- 3 hours), whereas proliferation of B cells in bone marrow, lymph node, and blood remained unchanged. Thus, the presence of one organ is necessary for appropriate expression of LFA-1 and ICAM-1 on B cells in other, distant organs. The more rapid transit of ICAM-1(low) B cells through lymph nodes may be responsible for the increased B-cell number in the blood after splenectomy.


Assuntos
Subpopulações de Linfócitos B/metabolismo , Molécula 1 de Adesão Intercelular/biossíntese , Antígeno-1 Associado à Função Linfocitária/biossíntese , Linfocitose/etiologia , Esplenectomia/efeitos adversos , Animais , Antígenos CD/biossíntese , Antígenos CD/genética , Subpopulações de Linfócitos B/imunologia , Células Sanguíneas/imunologia , Células Sanguíneas/metabolismo , Adesão Celular , Divisão Celular , Movimento Celular , Regulação para Baixo , Endotélio Vascular/citologia , Citometria de Fluxo , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/genética , Receptores de Hialuronatos/biossíntese , Receptores de Hialuronatos/genética , Imunofenotipagem , Integrina alfa4 , Molécula 1 de Adesão Intercelular/genética , Selectina L/biossíntese , Selectina L/genética , Linfonodos/citologia , Ativação Linfocitária , Contagem de Linfócitos , Antígeno-1 Associado à Função Linfocitária/genética , Especificidade de Órgãos , Ratos , Ratos Endogâmicos Lew , Receptores de Interleucina-2/biossíntese , Receptores de Interleucina-2/genética , Fatores de Tempo
13.
J Biol Chem ; 276(49): 45686-93, 2001 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-11571308

RESUMO

The inducible costimulator receptor (ICOS) is a third member of the CD28 receptor family that regulates T cell activation and function. ICOS binds to a newly identified ligand on antigen presenting cells different from the CD152 ligands CD80 and CD86. We used soluble ICOSIg and a newly developed murine anti-human ICOS ligand (ICOSL) monoclonal antibody to further characterize the ICOSL during ontogeny of antigen presenting cells. In a previous study, we found that ICOSL is expressed on monocytes, dendritic cells, and B cells. To define when ICOSL is first expressed on myeloid antigen presenting cells, we examined ICOSL expression on CD34(+) cells in bone marrow. We found that CD34(bright) cells regardless of their myeloid commitment were ICOSL(-), whereas ICOSL was first expressed when CD34 expression diminished and the myeloid marker CD33 appeared. However, acute myeloid leukemia cells were ICOSL-negative, whereas among B-cell malignancies only some cases of the most mature tumors such as prolymphocytic leukemia and hairy cell leukemia were positive. Next, we investigated purified CD34(+) hematopoietic progenitor cells that did not constitutively express ICOSL but were induced to express ICOSL within 12 h after granulocyte/macrophage colony-stimulating factor/tumor necrosis factor alpha (TNF-alpha) stimulation. Interestingly, ICOSL was induced prior to CD80/CD86 induction on CD34(+) cells so that ICOSL was expressed in the absence of CD80/CD86. This suggests that ICOSL is an early differentiation marker along the monocytic/dendritic maturation pathway. Induction of ICOSL was dependent on TNF-alpha and was regulated via NF-kappa B as revealed by use of inhibitors specific for I kappa B alpha phosphorylation such as BAY 11-7082 and BAY 11-7085. The antigen presenting capacity of TNF-alpha stimulated CD34(+) cells was strongly inhibited by ICOSIg fusion proteins or by NF-kappa B inhibition. Thus, TNF-alpha-induced ICOSL expression seemed to be functionally important for the costimulatory capacity of CD34(+) hematopoietic progenitor cells.


Assuntos
Antígenos CD34/imunologia , Células Dendríticas/imunologia , Regulação da Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Sequência de Bases , Diferenciação Celular , Primers do DNA , Células Dendríticas/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Ligantes , Células U937
14.
Laryngoscope ; 111(9): 1645-51, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11568621

RESUMO

OBJECTIVES: Acute laryngotracheitis is a disease in which mainly the subglottic area is infected, whereas adjacent parts of the larynx, especially the narrow glottic fold, remain unaffected. The reason for the difference between these two directly adjacent regions is unknown. Therefore, in the present study the influx of dendritic cells, neutrophils, T and B lymphocytes, natural killer cells, and macrophages into the mucosa of different laryngeal compartments was investigated after Sendai virus infection in the rat. The aims were to study both the influx of immunocompetent cells and the adhesion of the pathogen and to correlate them to the different reactions of the laryngeal areas during pseudocroup. METHODS: Acute laryngotracheitis was induced by intranasal application of Sendai virus in brown Norway rats. This virus is exclusively pneumotropic in rodents and belongs to the parainfluenza virus type 1, the main pathogen of acute laryngotracheitis in children. The numbers of dendritic cells, neutrophils, T and B lymphocytes, natural killer cells, and macrophages were determined in the supraglottic, glottic, subglottic, and tracheal mucosa on days 2, 5, 7, and 14 after virus application. Furthermore, the nucleoprotein of the virus and major histocompatibility complex (MHC) Class II expression were detected immunohistologically on the laryngeal epithelium. RESULTS: All cell subsets entered the laryngeal mucosa during inflammation. The highest influx was detected among dendritic cells subglottically. This was accompanied by a strong virus adhesion and MHC Class II expression on the subglottic epithelium. In contrast, only a few immunocompetent cells entered the adjacent glottic mucosa, and on the glottic epithelium staining for virus nucleoprotein and MHC Class II expression was weak. CONCLUSIONS: The inflammatory response of the laryngeal mucosa shows great regional differences in this animal model during experimental viral infection. The response was characterized by a strong subglottic and a weak glottic reaction. A possible reason for this difference might be region-specific viral adhesion on the epithelium of the laryngeal areas, as well as differences in MHC Class II expression. Thus, these data agree with the clinical observation during acute laryngotracheitis and may explain why the subglottic part of the larynx is affected preferentially during pseudocroup. The molecular mechanisms mediating the different reactions await clarification.


Assuntos
Modelos Animais de Doenças , Glote/imunologia , Imunocompetência/imunologia , Mucosa Laríngea/imunologia , Laringite/imunologia , Laringite/virologia , Infecções por Respirovirus/complicações , Respirovirus , Traqueíte/imunologia , Traqueíte/virologia , Doença Aguda , Animais , Linfócitos B/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Regulação Viral da Expressão Gênica/imunologia , Genes MHC da Classe II/imunologia , Glote/citologia , Imunidade Celular/imunologia , Imunidade nas Mucosas/imunologia , Imuno-Histoquímica , Células Matadoras Naturais/imunologia , Mucosa Laríngea/citologia , Contagem de Leucócitos , Macrófagos/imunologia , Neutrófilos/imunologia , Ratos , Linfócitos T/imunologia
15.
Cancer Immunol Immunother ; 49(11): 613-20, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11225992

RESUMO

Granulocyte/macrophage-colony-stimulating factor (GM-CSF) plays a central role in the differentiation and function of dendritic cells, which are crucial for the elicitation of MHC-restricted T cell responses. Preclinical and the first clinical data provide a rationale for the application of GM-CSF in immunotherapy of cancer. Ten patients with renal cell carcinoma stage IV (Holland/ Robson) were treated in this pilot study. Therapy was started with GM-CSF alone (2 weeks). Interleukin (IL-2) and interferon alpha (IFNalpha) were added sequentially (3 weeks GM-CSF plus IL-2 or IFNalpha, 3 weeks GM-CSF plus IL-2 plus IFNalpha). Therapy was performed on an outpatient basis. The cytokine regimen was evaluated for toxicity, clinical response and immunomodulatory effects [fluorescence-activated cell sorting analysis of peripheral blood mononuclear cells (PBMC), mixed-lymphocyte reaction and cytotoxicity of PBMC]. GM-CSF treatment caused a significant increase in the number of PBMC expressing costimulatory molecules. Addition of IL-2 and IFNalpha led to an increase in CD3 , CD4+, CD8+ and CD56+ PBMC in week 9. In an autologous mixed-lymphocyte reaction a 2.1-fold increase in T cell proliferation was observed after 2 weeks of GM-CSF treatment, and cytotoxicity assays showed changes in natural-killer-(NK)- and non-NK-mediated cytotoxicity in some patients. Two patients achieved partial remission, one patient had a mixed response. The toxicity of the regimen was mild to moderate with fever, flu-like symptoms and nausea being observed in most patients. Severe organ toxicity was not observed. We conclude that GM-CSF might be useful for immunotherapy of renal cell carcinoma, especially in combination with T-cell-active cytokines. Further studies are warranted.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Interferon-alfa/uso terapêutico , Interleucina-2/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Adolescente , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carcinoma de Células Renais/imunologia , Testes Imunológicos de Citotoxicidade , Esquema de Medicação , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos adversos , Humanos , Interferon-alfa/administração & dosagem , Interferon-alfa/efeitos adversos , Interleucina-2/administração & dosagem , Interleucina-2/efeitos adversos , Neoplasias Renais/imunologia , Contagem de Leucócitos , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Subpopulações de Linfócitos/classificação , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Projetos Piloto , Células Tumorais Cultivadas
16.
Infect Immun ; 68(12): 6704-11, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11083785

RESUMO

Viruses can escape T-cell surveillance by infecting macrophages and thereby induce apoptosis of noninfected T cells. This ability had not been demonstrated for bacteria. We investigated whether infection of macrophages with the important human pathogen Chlamydia trachomatis can induce T-cell apoptosis. Because Chlamydia-Mycoplasma coinfection is a frequent event, the ability of Mycoplasma fermentans-infected macrophages to induce T-cell apoptosis was also studied. Infected macrophages were cocultivated with autologous T cells in different activation states. Propidium iodide-based fluorescence-activated cell sorter analysis demonstrated that macrophages infected with viable chlamydiae induced T-cell death. Apoptosis was identified as the mode of death induction by using a terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling assay. Induction of T-cell death was macrophage dependent. Incubation of T cells with infectious chlamydiae in the absence of macrophages did not lead to T-cell apoptosis. UV irradiation of chlamydiae diminished the ability to induce death. T-cell death was induced by a cell-free supernatant of infected macrophages. Not only phytohemagglutinin-preactivated T cells but also non-mitogen-preactivated T cells were susceptible to C. trachomatis-induced apoptosis. In contrast, M. fermentans infection of macrophages did not induce T-cell death. Coinfection had no additional effect. In summary, intracellular chlamydial infection of macrophages can induce T-cell apoptosis. Apoptosis induction by chlamydiae possibly explains how persistently infected macrophages escape T-cell surveillance and why the Chlamydia-specific T-cell response is diminished during persistent chlamydial infection.


Assuntos
Apoptose , Chlamydia trachomatis/patogenicidade , Macrófagos/microbiologia , Linfócitos T/fisiologia , Chlamydia trachomatis/efeitos da radiação , Humanos , Ativação Linfocitária , Macrófagos/fisiologia , Fito-Hemaglutininas/farmacologia , Raios Ultravioleta
17.
J Clin Invest ; 106(5): 671-9, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10974020

RESUMO

This study used naked DNA vaccination to induce breakdown of tolerance to self and thus elicit immunological memory to native, membrane-bound Fas ligand (FasL). Upon induction of experimental autoimmune encephalomyelitis (EAE), this memory was turned on to provide protective immunity. FasL-specific autoantibodies isolated from protected animals differentially downregulated the in vitro production of TNF-alpha, but not IFN-gamma, by cultured T cells. These autoantibodies were highly protective when they were administered to rats at the onset of EAE. In contrast, administration of these FasL-specific Ab's to EAE rats after the peak of the acute phase of disease prevented spontaneous recovery from disease. This extended illness is partially explained by inhibition of mononuclear cell apoptosis at the target organ, which resulted in increased accumulation of T cells and macrophages at the site of inflammation. Hence, FasL exerts two distinct, stage-specific regulatory functions in the control of this T-cell mediated autoimmune disease of the central nervous system.


Assuntos
Autoanticorpos/imunologia , Encefalomielite Autoimune Experimental/imunologia , Glicoproteínas de Membrana/imunologia , Vacinas de DNA/imunologia , Animais , Apoptose , Encefalomielite Autoimune Experimental/terapia , Proteína Ligante Fas , Feminino , Imunidade Inata , Memória Imunológica , Imunoterapia , Interferon gama/biossíntese , Glicoproteínas de Membrana/uso terapêutico , Ratos , Ratos Endogâmicos Lew , Medula Espinal/patologia , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Vacinas de DNA/uso terapêutico
18.
Bone Marrow Transplant ; 25 Suppl 2: S46-9, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10933188

RESUMO

In chronic myeloid leukemia (CML) ex vivo generated DC are characterized by constitutive expression of bcr/abl and possibly other yet undefined leukemia-associated antigens, since these DC share a common progeny with leukemic cells. Induction of anti-leukemic T cell responses has been described in vitro. For a phase I vaccination study, autologous bcr/abl+ DC are generated under GMP conditions mainly from monocyte precursors in chronic phase CML patients. Lin-, CD80+, CD86+, CD83+, DR+ DC could be generated in sufficient numbers for s.c. vaccination with 1 x 10(6)-5 x 10(7) DC. Using monocyte precursors, the yield of DC per seeded PBMC was in the range of 1-6%. Furthermore, we could demonstrate in vitro that the T cell stimulatory ability of CD34+-derived DC can be augmented by a factor 2-3 by retroviral transduction with a gene coding for interleukin-7. DC-based vaccination strategies are a promising clinical approach, particularly as postremission immunotherapy in the setting of autologous stem cell transplantation.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/transplante , Proteínas de Fusão bcr-abl/imunologia , Leucemia Mieloide de Fase Crônica/terapia , Citotoxicidade Imunológica , Humanos , Interleucina-7/genética , Leucemia Mieloide de Fase Crônica/imunologia , Monócitos/imunologia , Transdução Genética , Transplante Autólogo , Vacinação
19.
Circulation ; 101(10): 1172-8, 2000 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-10715265

RESUMO

BACKGROUND: Fas (APO-1/CD95) is a transmembrane receptor belonging to the tumor necrosis factor receptor superfamily. Cross-linking of Fas by Fas ligand (FasL), a tumor necrosis factor-alpha-related cytokine, promotes apoptosis and/or transcription factor activation in a highly cell-type-specific manner. The biological consequences of Fas activation in cardiomyocytes and the regulation of Fas and FasL abundance in the myocardium in vivo remain largely unknown. METHODS AND RESULTS: As shown by immunohistochemistry, Fas was expressed on the sarcolemma of cardiomyocytes in left ventricular tissue sections. Moreover, FasL was constitutively expressed in the myocardium and in isolated cardiomyocytes, as revealed by reverse transcription polymerase chain reaction and Western blotting. Left ventricular abundance of Fas but not FasL was upregulated in a rat model of compensated volume-overload hypertrophy and was closely related to diastolic but not systolic wall stress as determined by MRI. Cardiomyocyte apoptosis was not enhanced in volume-overload hypertrophy despite the increased expression of Fas and the presence of FasL in the myocardium. Moreover, injection of mice with an agonistic anti-Fas antibody promoted hepatocyte but not cardiomyocyte apoptosis in vivo. Stimulation of isolated cardiomyocytes with recombinant FasL promoted an activation of the transcription factor AP-1 as shown by electrophoretic mobility shift assays but did not induce cell death. CONCLUSIONS: Fas and FasL are constitutively expressed in the myocardium and in cardiomyocytes. Myocardial expression of Fas is closely related to diastolic loading conditions in vivo. Signaling pathways emanating from Fas are coupled to an activation of the transcription factor AP-1 in cardiomyocytes.


Assuntos
Cardiomegalia/metabolismo , Glicoproteínas de Membrana/biossíntese , Miocárdio/metabolismo , Fator de Transcrição AP-1/metabolismo , Receptor fas/biossíntese , Animais , Apoptose , Cardiomegalia/patologia , Sobrevivência Celular , Proteína Ligante Fas , Masculino , Camundongos , NF-kappa B/metabolismo , Ratos , Ratos Endogâmicos WKY , Transdução de Sinais , Função Ventricular Esquerda/fisiologia
20.
Exp Hematol ; 27(9): 1402-8, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10480431

RESUMO

T-cell apoptosis is a mechanism regulating T-cell homeostasis. Activation renders T cells susceptible to activation-induced cell death, a process mediated through CD95 ligand/CD95 (Apo-1/Fas) ligation. The aim of this study was to test whether antigen-presenting cells can inhibit CD95/Fas-triggered activation-induced cell death. Dendritic cells (DC), which are highly effective antigen-presenting cells, were generated in vitro from human peripheral blood monocytes by culture in granulocyte-macrophage colony-stimulating factor and interleukin 4. Subsequently, DC were cocultured with activated T cells and the effect of DC on CD95/Fas-mediated apoptosis was determined. Coculture with increasing amounts of DC prevented CD95/Fas-triggered apoptosis in a dose-dependent fashion by inhibiting activation of caspase 8 and caspase 3. This protective effect of the DC on T-cell death could be blocked by 50% by adding an anti-CD58 antibody, whereas further addition of anti-CD80 (B7.1) and anti-CD86 (B7.2) led to an even more pronounced effect. Our findings suggest that DC can protect T cells from activation-induced cell death, with CD58 ligation playing a key role.


Assuntos
Apresentação de Antígeno , Apoptose , Antígenos CD58/fisiologia , Células Dendríticas/fisiologia , Ativação Linfocitária , Linfócitos T/citologia , Receptor fas/fisiologia , Anticorpos Monoclonais/farmacologia , Antígenos CD/fisiologia , Antígeno B7-1/fisiologia , Antígeno B7-2 , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/fisiologia , Células Cultivadas , Técnicas de Cocultura , Relação Dose-Resposta Imunológica , Ativação Enzimática , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interleucina-4/farmacologia , Glicoproteínas de Membrana/fisiologia , Monócitos/efeitos dos fármacos , Linfócitos T/enzimologia , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA