Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Bone Marrow Transplant ; 57(6): 911-917, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35368040

RESUMO

Acute Myeloid Leukemia (AML) has a median age at diagnosis of 67 years. The most common curative therapy remains an allogeneic hematopoietic stem cell transplantation (HCT), yet it is complicated by treatment-related mortality (TRM) and ongoing morbidity including graft versus host disease (GVHD) that may impact survival, particularly in older patients. We examined the outcomes and predictors of success in 1321 patients aged 60 years and older receiving a HCT for AML in first complete remission (CR1) from 2007-2017 and reported to the CIBMTR. Outcomes were compared in three age cohorts (60-64; 65-69; 70+). With median follow-up of nearly 3 years, patients aged 60-64 had modestly, though significantly better OS, DFS and lower TRM than those either 65-69 or 70+; cohorts with similar outcomes. Three-year OS for the 3 cohorts was 49.4%, 42.3%, and 44.7% respectively (p = 0.026). TRM was higher with increasing age, cord blood as graft source and HCT-CI score of ≥3. Conditioning intensity was not a significant predictor of OS in the 60-69 cohort with 3-year OS of 46% for RIC and 49% for MAC (p = 0.38); MAC was rarely used over age 70. There was no difference in the relapse rate, incidence of Grade III/IV acute GVHD, or moderate-severe chronic GVHD across the age cohorts. After adjusting for other predictors, age had a small effect on OS and TRM. High-risk features including poor cytogenetics and measurable residual disease (MRD) prior to HCT were each significantly associated with relapse and accounted for most of the adverse impact on OS and DFS. Age did not influence the incidence of either acute or chronic GVHD; while graft type and associated GVHD prophylaxis were most important. These data suggest that age alone is not a barrier to successful HCT for AML in CR1 and should not exclude patients from HCT. Efforts should focus on minimizing residual disease and better donor selection.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Leucemia Mieloide Aguda , Adulto , Idoso , Doença Enxerto-Hospedeiro/etiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Pessoa de Meia-Idade , Neoplasia Residual , Receptores de Complemento 3b/uso terapêutico , Recidiva , Estudos Retrospectivos , Condicionamento Pré-Transplante/efeitos adversos
2.
Adv Chronic Kidney Dis ; 21(2): 152-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24602464

RESUMO

A major shift in our understanding of glomerular diseases is the focus on which components of the complement pathway are involved in mediating kidney injury. For example, the membranoproliferative glomerulonephritis lesion is no longer classified solely by ultrastructural findings on biopsy and is now divided into immune-complex-mediated lesions vs complement-mediated lesions. In turn, this emphasis on complement leads to interest in therapies that target complement as potential disease-modifying agents. Eculizumab, the first available anti-complement therapy, blocks at the level of C5 and has revolutionized the treatment of atypical hemolytic uremic syndrome. Whether this agent will work equally well for the far more heterogeneous entities of C3 glomerulonephritis and dense deposit disease remains unclear. Instead, newer agents that target C3 may turn out to be the most effective and specific therapy for these C3 glomerulopathies.


Assuntos
Inativadores do Complemento/uso terapêutico , Proteínas do Sistema Complemento/imunologia , Glomerulonefrite Membranoproliferativa/tratamento farmacológico , Síndrome Hemolítico-Urêmica/tratamento farmacológico , Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica , Complemento C3/antagonistas & inibidores , Complemento C3/imunologia , Complemento C5/antagonistas & inibidores , Complemento C5/imunologia , Glomerulonefrite/tratamento farmacológico , Glomerulonefrite/imunologia , Glomerulonefrite Membranoproliferativa/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Humanos , Receptores de Complemento 3b/uso terapêutico
3.
Eur J Immunol ; 41(11): 3301-11, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21830207

RESUMO

Although the exact etiology of systemic lupus erythematosus (SLE) remains elusive, B-cell hyperactivity and production of autoantibodies directed to components of the cell nucleus are a well-established pathogenetic mechanism of the disease. Therefore, the targeted inhibition of DNA-specific B cells is a logical therapeutic approach. The complement receptor type 1 (CR1, CD35) has been shown to suppress human B-cell activation and proliferation after co-cross-linking with the BCR, and may serve as a mediator for negative signal delivery. In order to evaluate this therapeutic approach in a human-like system, we used immune-restricted SCID mice transferred with PBMCs from SLE patients. The tolerance of these humanized SCID mice to native DNA was re-established after administration of a chimeric molecule consisting of a CR1-specific mAb coupled to the decapeptide DWEYSVWLSN that mimics dsDNA. The generated protein-engineered chimera was able to co-cross-link selectively native DNA-specific BCR with the B-cell inhibitory receptor CR1, thus delivering a strong inhibitory signal.


Assuntos
Anticorpos Antinucleares/imunologia , Anticorpos Monoclonais/imunologia , Linfócitos B/imunologia , Imunoterapia/métodos , Lúpus Eritematoso Sistêmico/imunologia , Animais , Anticorpos Antinucleares/biossíntese , Anticorpos Antinucleares/uso terapêutico , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/uso terapêutico , Autoantígenos/imunologia , Autoimunidade/imunologia , Western Blotting , Linhagem Celular , Separação Celular , DNA/imunologia , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Imunoprecipitação , Ativação Linfocitária/imunologia , Camundongos , Camundongos SCID , Peptídeos , Receptores de Complemento 3b/imunologia , Receptores de Complemento 3b/uso terapêutico , Transdução de Sinais/imunologia
4.
J Biol Chem ; 284(51): 35605-11, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19833734

RESUMO

CRIg is a recently discovered complement C3 receptor expressed on a subpopulation of tissue-resident macrophages. The extracellular IgV domain of CRIg (CRIg-ECD) holds considerable promise as a potential therapeutic because it selectively inhibits the alternative pathway of complement by binding to C3b and inhibiting proteolytic activation of C3 and C5. However, CRIg binds weakly to the convertase subunit C3b (K(D) = 1.1 microm), and thus a relatively high concentration of protein is required to reach nearly complete complement inhibition. To improve therapeutic efficacy while minimizing risk of immunogenicity, we devised a phage display strategy to evolve a high affinity CRIg-ECD variant with a minimal number of mutations. Using the crystal structure of CRIg in complex with C3b as a guide for library design, we isolated a CRIg-ECD double mutant (Q64R/M86Y, CRIg-v27) that showed increased binding affinity and improved complement inhibitory activity relative to CRIg-ECD. In a mouse model of arthritis, treatment with a Fc fusion of CRIg-v27 resulted in a significant reduction in clinical scores compared with treatment with an Fc fusion of CRIg-ECD. This study clearly illustrates how phage display technology and structural information can be combined to generate proteins with nearly natural sequences that act as potent complement inhibitors with greatly improved therapeutic efficacy.


Assuntos
Artrite/tratamento farmacológico , Receptores de Complemento 3b/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Substituição de Aminoácidos , Animais , Artrite/metabolismo , Complemento C3b/genética , Complemento C3b/metabolismo , Complemento C5/genética , Complemento C5/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mutação de Sentido Incorreto , Estrutura Terciária de Proteína/fisiologia , Coelhos , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Relação Estrutura-Atividade
5.
Immunol Rev ; 180: 177-89, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11414360

RESUMO

With increasing evidence that complement activation significantly contributes to the pathogenesis of a large number of inflammatory diseases, strategies that interfere with its deleterious action have become a major focus in pharmacological research. Endogenous soluble complement inhibitors (C1 inhibitor, recombinant soluble complement receptor 1, antibodies) blocking key proteins of the cascade reaction, neutralizing the action of the complement-derived anaphylatoxin C5a, or interfering with complement receptor 3 (CR3, CD18/11b)-mediated adhesion of inflammatory cells to the vascular endothelium have successfully been tested in various animal models over the past years. Promising results consequently led to clinical trials. Furthermore, incorporation of membrane-bound complement regulators (decay-accelerating factor (CD55), membrane co-factor protein (CD46), CD59) in transgenic animals has provided a major step forward in protecting xenografts from hyperacute rejection. At the same time, the poor contribution of complement to the antitumor response, which is caused by multiple resistance mechanisms that hamper the efficacy of antibody-based tumor therapy, is increasingly recognized and requires pharmacologic intervention. First attempts have now been made to interfere with the resistance mechanisms, thereby improving complement-mediated tumor cell destruction.


Assuntos
Anti-Inflamatórios/uso terapêutico , Ativação do Complemento/efeitos dos fármacos , Proteínas Inativadoras do Complemento/uso terapêutico , Proteínas do Sistema Complemento/fisiologia , Desenho de Fármacos , Anafilatoxinas/antagonistas & inibidores , Anafilatoxinas/imunologia , Angioedema/tratamento farmacológico , Animais , Animais Geneticamente Modificados , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Anticorpos Antineoplásicos/uso terapêutico , Benzamidinas , Proteínas Inativadoras do Complemento 1/uso terapêutico , Proteínas Inativadoras do Complemento/química , Proteínas Inativadoras do Complemento/farmacologia , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/química , Cães , Avaliação Pré-Clínica de Medicamentos , Rejeição de Enxerto/prevenção & controle , Guanidinas/uso terapêutico , Humanos , Imunoterapia , Inflamação/imunologia , Inflamação/prevenção & controle , Macaca fascicularis , Camundongos , Neoplasias/imunologia , Neoplasias/terapia , Pancreatite/tratamento farmacológico , Coelhos , Ratos , Receptores de Complemento/efeitos dos fármacos , Receptores de Complemento 3b/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Suínos , Doadores de Tecidos , Transfecção , Transplante Heterólogo
6.
J Immunol ; 162(8): 4952-9, 1999 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10202042

RESUMO

The complement inhibitor soluble complement receptor type 1 (sCR1) and a truncated form of sCR1, sCR1[desLHR-A], have been generated with expression of the selectin-reactive oligosaccharide moiety, sialyl Lewisx (sLex), as N-linked oligosaccharide adducts. These modified proteins, sCR1sLex and sCR1[desLHR-A]sLex, were assessed in the L-selectin- and P-selectin-dependent rat model of lung injury following systemic activation of complement by cobra venom factor and in the L-selectin-, P-selectin-, and E-selectin-dependent model of lung injury following intrapulmonary deposition of IgG immune complexes. In the cobra venom factor model, sCR1sLex and sCR1[desLHR-A]sLex caused substantially greater reductions in neutrophil accumulation and in albumin extravasation in lung when compared with the non-sLex-decorated forms. In this model, increased lung vascular binding of sCR1sLex and sCR1[desLHR-A]sLex occurred in a P-selectin-dependent manner, in contrast to the absence of any increased binding of sCR1 or sCR1[desLHR-A]. In the IgG immune complex model, sCR1[desLHR-A]sLex possessed greater protective effects relative to sCR1[desLHR-A], based on albumin extravasation and neutrophil accumulation. Enhanced protective effects correlated with greater lung vascular binding of sCR1[desLHR-A]sLex as compared with the non-sLex-decorated form. In TNF-alpha-activated HUVEC, substantial in vitro binding occurred with sCR1[desLHR-A]sLex (but not with sCR1[desLHR-A]). This endothelial cell binding was blocked by anti-E-selectin but not by anti-P-selectin. These data suggest that sLex-decorated complement inhibitors have enhanced antiinflammatory effects and appear to have enhanced ability to localize to the activated vascular endothelium.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Proteínas Inativadoras do Complemento/uso terapêutico , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Pulmão/patologia , Oligossacarídeos/imunologia , Anti-Inflamatórios não Esteroides/imunologia , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/imunologia , Venenos Elapídicos/administração & dosagem , Endotélio Vascular/metabolismo , Humanos , Doenças do Complexo Imune/imunologia , Doenças do Complexo Imune/patologia , Doenças do Complexo Imune/terapia , Imuno-Histoquímica , Infusões Intravenosas , Antígenos do Grupo Sanguíneo de Lewis/genética , Pulmão/irrigação sanguínea , Pulmão/química , Pulmão/metabolismo , Oligossacarídeos/genética , Oligossacarídeos/uso terapêutico , Ligação Proteica/imunologia , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/uso terapêutico , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico , Sequências Repetitivas de Aminoácidos , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Antígeno Sialil Lewis X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA