Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
N Engl J Med ; 376(5): 429-439, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-27959701

RESUMO

BACKGROUND: The up-regulation of P-selectin in endothelial cells and platelets contributes to the cell-cell interactions that are involved in the pathogenesis of vaso-occlusion and sickle cell-related pain crises. The safety and efficacy of crizanlizumab, an antibody against the adhesion molecule P-selectin, were evaluated in patients with sickle cell disease. METHODS: In this double-blind, randomized, placebo-controlled, phase 2 trial, we assigned patients to receive low-dose crizanlizumab (2.5 mg per kilogram of body weight), high-dose crizanlizumab (5.0 mg per kilogram), or placebo, administered intravenously 14 times over a period of 52 weeks. Patients who were receiving concomitant hydroxyurea as well as those not receiving hydroxyurea were included in the study. The primary end point was the annual rate of sickle cell-related pain crises with high-dose crizanlizumab versus placebo. The annual rate of days hospitalized, the times to first and second crises, annual rates of uncomplicated crises (defined as crises other than the acute chest syndrome, hepatic sequestration, splenic sequestration, or priapism) and the acute chest syndrome, and patient-reported outcomes were also assessed. RESULTS: A total of 198 patients underwent randomization at 60 sites. The median rate of crises per year was 1.63 with high-dose crizanlizumab versus 2.98 with placebo (indicating a 45.3% lower rate with high-dose crizanlizumab, P=0.01). The median time to the first crisis was significantly longer with high-dose crizanlizumab than with placebo (4.07 vs. 1.38 months, P=0.001), as was the median time to the second crisis (10.32 vs. 5.09 months, P=0.02). The median rate of uncomplicated crises per year was 1.08 with high-dose crizanlizumab, as compared with 2.91 with placebo (indicating a 62.9% lower rate with high-dose crizanlizumab, P=0.02). Adverse events that occurred in 10% or more of the patients in either active-treatment group and at a frequency that was at least twice as high as that in the placebo group were arthralgia, diarrhea, pruritus, vomiting, and chest pain. CONCLUSIONS: In patients with sickle cell disease, crizanlizumab therapy resulted in a significantly lower rate of sickle cell-related pain crises than placebo and was associated with a low incidence of adverse events. (Funded by Selexys Pharmaceuticals and others; SUSTAIN ClinicalTrials.gov number, NCT01895361 .).


Assuntos
Anemia Falciforme/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Selectina-P/antagonistas & inibidores , Dor/prevenção & controle , Adolescente , Adulto , Anemia Falciforme/complicações , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Humanos , Hidroxiureia/uso terapêutico , Masculino , Pessoa de Meia-Idade , Selectina-P/imunologia , Dor/etiologia , Qualidade de Vida , Adulto Jovem
2.
Circ Res ; 104(4): 550-8, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19131645

RESUMO

Complement is a central effector system within the immune system and is implicated in a range of inflammatory disorders. CD59 is a key regulator of complement membrane attack complex (MAC) assembly. The atherogenic role of terminal complement has long been suspected but is still unclear. Here, we demonstrate that among mice deficient in apolipoprotein (Apo)E, the additional loss of murine CD59 (mCd59ab(-/-)/ApoE(-/-)) accelerated advanced atherosclerosis featuring occlusive coronary atherosclerosis, vulnerable plaque, and premature death and that these effect could be attenuated by overexpression of human CD59 in the endothelium. Complement inhibition using a neutralizing anti-mouse C5 antibody attenuated atherosclerosis in mCd59ab(-/-)/ApoE(-/-) mice. Furthermore, MAC mediated endothelial damage and promoted foam cell formation. These combined results highlight the atherogenic role of MAC and the atheroprotective role of CD59 and suggest that inhibition of MAC formation may provide a therapeutic approach for the treatment of atherosclerosis.


Assuntos
Aterosclerose/prevenção & controle , Antígenos CD59/metabolismo , Ativação do Complemento , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/patologia , Antígenos CD59/genética , Linhagem Celular , Complemento C5/imunologia , Complemento C9/imunologia , Doença da Artéria Coronariana/imunologia , Doença da Artéria Coronariana/prevenção & controle , Gorduras na Dieta/administração & dosagem , Modelos Animais de Doenças , Progressão da Doença , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Células Espumosas/imunologia , Células Espumosas/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Coelhos , Índice de Gravidade de Doença , Fatores de Tempo
3.
Brain ; 133(Pt 7): 1944-60, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20513658

RESUMO

The motor axonal variant of Guillain-Barré syndrome is associated with anti-GD1a immunoglobulin antibodies, which are believed to be the pathogenic factor. In previous studies we have demonstrated the motor terminal to be a vulnerable site. Here we show both in vivo and ex vivo, that nodes of Ranvier in intramuscular motor nerve bundles are also targeted by anti-GD1a antibody in a gradient-dependent manner, with greatest vulnerability at distal nodes. Complement deposition is associated with prominent nodal injury as monitored with electrophysiological recordings and fluorescence microscopy. Complete loss of nodal protein staining, including voltage-gated sodium channels and ankyrin G, occurs and is completely protected by both complement and calpain inhibition, although the latter provides no protection against electrophysiological dysfunction. In ex vivo motor and sensory nerve trunk preparations, antibody deposits are only observed in experimentally desheathed nerves, which are thereby rendered susceptible to complement-dependent morphological disruption, nodal protein loss and reduced electrical activity of the axon. These studies provide a detailed mechanism by which loss of axonal conduction can occur in a distal dominant pattern as observed in a proportion of patients with motor axonal Guillain-Barré syndrome, and also provide an explanation for the occurrence of rapid recovery from complete paralysis and electrophysiological in-excitability. The study also identifies therapeutic approaches in which nodal architecture can be preserved.


Assuntos
Autoanticorpos/toxicidade , Calpaína/metabolismo , Ativação do Complemento/imunologia , Gangliosídeos/imunologia , Neurônios Motores/imunologia , Neurônios Motores/patologia , Nós Neurofibrosos/imunologia , Nós Neurofibrosos/patologia , Animais , Autoanticorpos/metabolismo , Axônios/imunologia , Axônios/patologia , Sítios de Ligação de Anticorpos , Síndrome de Guillain-Barré/imunologia , Síndrome de Guillain-Barré/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Camundongos Transgênicos , Neurônios Motores/metabolismo , Nós Neurofibrosos/metabolismo
4.
Br J Haematol ; 149(3): 414-25, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20230403

RESUMO

Pulmonary hypertension (PH) is a common complication of haemolytic anaemia. Intravascular haemolysis leads to nitric oxide (NO) depletion, endothelial and smooth muscle dysregulation, and vasculopathy, characterized by progressive hypertension. PH has been reported in patients with paroxysmal nocturnal haemoglobinuria (PNH), a life-threatening haemolytic disease. We explored the relationship between haemolysis, systemic NO, arginine catabolism and measures of PH in 73 PNH patients enrolled in the placebo-controlled TRIUMPH (Transfusion Reduction Efficacy and Safety Clinical Investigation Using Eculizumab in Paroxysmal Nocturnal Haemoglobinuria) study. At baseline, intravascular haemolysis was associated with elevated NO consumption (P < 0.0001) and arginase-1 release (P < 0.0001). Almost half of the patients in the trial had elevated levels (> or =160 pg/ml) of N-terminal pro-brain natriuretic peptide (NT-proBNP), a marker of pulmonary vascular resistance and right ventricular dysfunction previously shown to indicate PH. Eculizumab treatment significantly reduced haemolysis (P < 0.001), NO depletion (P < 0.001), vasomotor tone (P < 0.05), dyspnoea (P = 0.006) and resulted in a 50% reduction in the proportion of patients with elevated NT-proBNP (P < 0.001) within 2 weeks of treatment. Importantly, the significant improvements in dyspnoea and NT-proBNP levels occurred without significant changes in anaemia. These data demonstrated that intravascular haemolysis in PNH produces a state of NO catabolism leading to signs of PH, including elevated NT pro-BNP and dyspnoea that are significantly improved by treatment with eculizumab.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Dispneia/tratamento farmacológico , Hemoglobinúria Paroxística/tratamento farmacológico , Hemólise/efeitos dos fármacos , Hipertensão Pulmonar/tratamento farmacológico , Óxido Nítrico/deficiência , Anticorpos Monoclonais Humanizados , Biomarcadores/sangue , Pressão Sanguínea/efeitos dos fármacos , Dispneia/etiologia , Feminino , Hemoglobinas/metabolismo , Hemoglobinúria Paroxística/sangue , Hemoglobinúria Paroxística/complicações , Hemólise/fisiologia , Humanos , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/fisiopatologia , Masculino , Peptídeo Natriurético Encefálico/sangue , Fragmentos de Peptídeos/sangue , Resultado do Tratamento
5.
Br J Haematol ; 149(3): 446-50, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20151973

RESUMO

In Paroxysmal nocturnal haemoglobinuria (PNH), pregnancy is associated with increased maternal and foetal complications to such an extent that the condition has been considered relatively contra-indicated in PNH. Eculizumab has revolutionized the treatment of PNH. We evaluate its use in pregnancy to date. We report on seven patients exposed to eculizumab at different stages of pregnancy including the first two patients to receive the drug from conception to delivery. There was no evidence of complement blockade from cord blood samples taken at delivery. Eculizumab appears safe to use in this setting and is likely to prevent many of the complications usually observed.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Hemoglobinúria Paroxística/tratamento farmacológico , Complicações Hematológicas na Gravidez/tratamento farmacológico , Adulto , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais Humanizados , Feminino , Sangue Fetal/metabolismo , Humanos , Troca Materno-Fetal , Gravidez , Adulto Jovem
6.
Haematologica ; 95(4): 567-73, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20145265

RESUMO

BACKGROUND: Paroxysmal nocturnal hemoglobinuria is an acquired hemolytic anemia characterized by intravascular hemolysis which has been demonstrated to be effectively controlled with eculizumab. However, lactate dehydrogenase levels remain slightly elevated and haptoglobin levels remain low in some patients suggesting residual low-level hemolysis. This may be due to C3-mediated clearance of paroxysmal nocturnal hemoglobinuria red blood cells through the reticuloendothelial system. DESIGN AND METHODS: Thirty-nine samples from patients not treated with eculizumab and 31 samples from patients treated with eculizumab were obtained (for 17 of these 31 samples there were also samples taken prior to eculizumab treatment). Membrane bound complement was assessed by flow cytometry. Direct antiglobulin testing was carried out using two methods. Lactate dehydrogenase was assayed to assess the degree of hemolysis. RESULTS: Three of 39 patients (8%) with paroxysmal nocturnal hemoglobinuria not on eculizumab had a positive direct antiglobulin test, while the test was positive in 21 of 31 (68%) during eculizumab treatment. Of these 21 patients who had a positive direct antiglobulin test during eculizumab treatment, 17 had been tested prior to treatment; only one was positive. Flow cytometry using anti-C3 monoclonal antibodies was performed on the 21 direct antiglobulin test-positive, eculizumab-treated patients; the median proportion of C3-positive total red blood cells was 26%. Among the eculizumab-treated patients, 16 of the 21 (76.2%) with a positive direct antiglobulin test received at least one transfusion compared with one of ten (10.0%) of those with a negative test (P<0.01). Among the eculizumab-treated patients, the mean hemoglobin value for the 21 with a positive direct antiglobulin test was 9.6+/-0.3 g/dL, whereas that in the ten patients with a negative test was 11.0+/-0.4 g/dL (P=0.02). CONCLUSIONS: These data demonstrate a previously masked mechanism of red cell clearance in paroxysmal nocturnal hemoglobinuria and suggests that blockade of complement at C5 allows C3 fragment accumulation on some paroxysmal nocturnal hemoglobinuria red cells, explaining the residual low-level hemolysis occurring in some eculizumab-treated patients.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complemento C3/imunologia , Hemoglobinúria Paroxística/prevenção & controle , Hemólise/efeitos dos fármacos , Imunoterapia , Anticorpos Monoclonais Humanizados , Transfusão de Sangue , Complemento C5/antagonistas & inibidores , Complemento C5/imunologia , Eritrócitos/metabolismo , Citometria de Fluxo , Hemoglobinas/metabolismo , Hemoglobinúria Paroxística/sangue , Hemoglobinúria Paroxística/imunologia , Humanos , L-Lactato Desidrogenase/metabolismo
7.
Am J Hematol ; 85(8): 553-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20658586

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is a debilitating and life-threatening disease in which lysis of PNH red blood cells frequently manifests with chronic hemolysis, anemia, and thrombosis. Renal damage in PNH is associated with chronic hemosiderosis and/or microvascular thrombosis. We determined the incidence of renal dysfunction or damage, defined by stages of chronic kidney disease (CKD), in a large cohort of PNH patients and evaluated the safety and efficacy of the complement inhibitor eculizumab in altering its progression. Renal dysfunction or damage was observed in 65% of the study population at baseline with 21% of patients with later stage CKD or kidney failure (glomerular filtration rate [GFR]

Assuntos
Anticorpos Monoclonais/uso terapêutico , Complemento C5/antagonistas & inibidores , Inativadores do Complemento/uso terapêutico , Hemoglobinúria Paroxística/tratamento farmacológico , Falência Renal Crônica/prevenção & controle , Rim/fisiopatologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Ativação do Complemento/efeitos dos fármacos , Complemento C5/imunologia , Feminino , Taxa de Filtração Glomerular/efeitos dos fármacos , Hemoglobinúria Paroxística/complicações , Hemoglobinúria Paroxística/imunologia , Hemoglobinúria Paroxística/fisiopatologia , Hemólise/efeitos dos fármacos , Humanos , Rim/efeitos dos fármacos , Falência Renal Crônica/tratamento farmacológico , Falência Renal Crônica/etiologia , Falência Renal Crônica/fisiopatologia , Masculino , Taxa de Depuração Metabólica/efeitos dos fármacos , Pessoa de Meia-Idade , Projetos Piloto , Índice de Gravidade de Doença , Resultado do Tratamento , Adulto Jovem
8.
Nat Biotechnol ; 25(11): 1256-64, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17989688

RESUMO

The complement system provides critical immunoprotective and immunoregulatory functions but uncontrolled complement activation can lead to severe pathology. In the rare hemolytic disease paroxysmal nocturnal hemoglobinuria (PNH), somatic mutations result in a deficiency of glycosylphosphatidylinositol-linked surface proteins, including the terminal complement inhibitor CD59, on hematopoietic stem cells. In a dysfunctional bone marrow background, these mutated progenitor blood cells expand and populate the periphery. Deficiency of CD59 on PNH red blood cells results in chronic complement-mediated intravascular hemolysis, a process central to the morbidity and mortality of PNH. A recently developed, humanized monoclonal antibody directed against complement component C5, eculizumab (Soliris; Alexion Pharmaceuticals Inc., Cheshire, CT, USA), blocks the proinflammatory and cytolytic effects of terminal complement activation. The recent approval of eculizumab as a first-in-class complement inhibitor for the treatment of PNH validates the concept of complement inhibition as an effective therapy and provides rationale for investigation of other indications in which complement plays a role.


Assuntos
Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Complemento C5/antagonistas & inibidores , Inativadores do Complemento/química , Inativadores do Complemento/farmacologia , Desenho de Fármacos , Hemoglobinúria Paroxística/tratamento farmacológico , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Ensaios Clínicos como Assunto , Complemento C5/fisiologia , Inativadores do Complemento/uso terapêutico , Aprovação de Drogas , Avaliação Pré-Clínica de Medicamentos , Hemoglobinúria Paroxística/imunologia , Humanos , Camundongos , Engenharia de Proteínas
9.
N Engl J Med ; 355(12): 1233-43, 2006 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-16990386

RESUMO

BACKGROUND: We tested the safety and efficacy of eculizumab, a humanized monoclonal antibody against terminal complement protein C5 that inhibits terminal complement activation, in patients with paroxysmal nocturnal hemoglobinuria (PNH). METHODS: We conducted a double-blind, randomized, placebo-controlled, multicenter, phase 3 trial. Patients received either placebo or eculizumab intravenously; eculizumab was given at a dose of 600 mg weekly for 4 weeks, followed 1 week later by a 900-mg dose and then 900 mg every other week through week 26. The two primary end points were the stabilization of hemoglobin levels and the number of units of packed red cells transfused. Biochemical indicators of intravascular hemolysis and the patients' quality of life were also assessed. RESULTS: Eighty-seven patients underwent randomization. Stabilization of hemoglobin levels in the absence of transfusions was achieved in 49% (21 of 43) of the patients assigned to eculizumab and none (0 of 44) of those assigned to placebo (P<0.001). During the study, a median of 0 units of packed red cells was administered in the eculizumab group, as compared with 10 units in the placebo group (P<0.001). Eculizumab reduced intravascular hemolysis, as shown by the 85.8% lower median area under the curve for lactate dehydrogenase plotted against time (in days) in the eculizumab group, as compared with the placebo group (58,587 vs. 411,822 U per liter; P<0.001). Clinically significant improvements were also found in the quality of life, as measured by scores on the Functional Assessment of Chronic Illness Therapy-Fatigue instrument (P<0.001) and the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire. Of the 87 patients, 4 in the eculizumab group and 9 in the placebo group had serious adverse events, none of which were considered to be treatment-related; all these patients recovered without sequelae. CONCLUSIONS: Eculizumab is an effective therapy for PNH.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complemento C5/imunologia , Hemoglobinúria Paroxística/tratamento farmacológico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais Humanizados , Método Duplo-Cego , Transfusão de Eritrócitos , Fadiga , Feminino , Hemoglobinas/análise , Hemoglobinúria Paroxística/sangue , Hemoglobinúria Paroxística/terapia , Hemólise/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Qualidade de Vida
10.
Liver Transpl ; 15(5): 540-3, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19399743

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare, acquired hemolytic anemia caused by somatic mutations in the phosphatidylinositol glycan-complementation class A gene and the resulting absence of a key complement regulatory protein, CD59. Affected red blood cells in patients with PNH undergo intravascular complement-mediated lysis with resulting anemia, hemoglobinuria, and venous thromboses. Hepatic venous outflow thrombosis [Budd-Chiari syndrome (BCS)] is especially common in PNH patients and often fatal. The few case reports of outcomes in patients undergoing liver transplant for BCS secondary to PNH detail instances of recurrent BCS as well as early thrombotic portal vein occlusion and hepatic artery thrombosis requiring retransplantation. PNH is therefore generally considered a contraindication to liver transplantation. Here we present the first report of a patient with PNH and BCS undergoing successful liver transplantation while receiving eculizumab, a humanized monoclonal antibody that blocks the activation of the terminal complement at C5.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Síndrome de Budd-Chiari/cirurgia , Complemento C5/antagonistas & inibidores , Hemoglobinúria Paroxística/tratamento farmacológico , Transplante de Fígado , Adolescente , Anticorpos Monoclonais Humanizados , Anticoagulantes/uso terapêutico , Síndrome de Budd-Chiari/sangue , Síndrome de Budd-Chiari/etiologia , Síndrome de Budd-Chiari/imunologia , Hemoglobinúria Paroxística/sangue , Hemoglobinúria Paroxística/complicações , Hemoglobinúria Paroxística/imunologia , Hemólise/efeitos dos fármacos , Artéria Hepática/cirurgia , Humanos , Veia Ilíaca/transplante , Imunossupressores/uso terapêutico , Masculino , Veias Mesentéricas/cirurgia , Veia Porta/cirurgia , Resultado do Tratamento , Veia Cava Inferior/cirurgia
11.
Brain ; 131(Pt 5): 1197-208, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18184663

RESUMO

Anti-GQ1b ganglioside antibodies are the serological hallmark of the Miller Fisher syndrome (MFS) variant of the paralytic neuropathy, Guillain-Barré syndrome, and are believed to be the principal pathogenic mediators of the disease. In support of this, we previously showed in an in vitro mouse model of MFS that anti-GQ1b antibodies were able to bind and disrupt presynaptic motor nerve terminals at the neuromuscular junction (NMJ) as one of their target sites, thereby causing muscle paralysis. This injury only occurred through activation of complement, culminating in the formation and deposition of membrane attack complex (MAC, C5b-9) in nerve membranes. Since this step is crucial to the neuropathic process and an important convergence point for antibody and complement mediated membrane injury in general, it forms an attractive pharmacotherapeutic target. Here, we assessed the efficacy of the humanized monoclonal antibody eculizumab, which blocks the formation of human C5a and C5b-9, in preventing the immune-mediated motor neuropathy exemplified in this model. Eculizumab completely prevented electrophysiological and structural lesions at anti-GQ1b antibody pre-incubated NMJs in vitro when using normal human serum (NHS) as a complement source. In a novel in vivo mouse model of MFS generated through intraperitoneal injection of anti-GQ1b antibody and NHS, mice developed respiratory paralysis due to transmission block at diaphragm NMJs, resulting from anti-GQ1b antibody binding and complement activation. Intravenous injection of eculizumab effectively prevented respiratory paralysis and associated functional and morphological hallmarks of terminal motor neuropathy. We show that eculizumab protects against complement-mediated damage in murine MFS, providing the rationale for undertaking clinical trials in this disease and other antibody-mediated neuropathies in which complement activation is believed to be involved.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Síndrome de Miller Fisher/prevenção & controle , Doença Autoimune do Sistema Nervoso Experimental/prevenção & controle , Animais , Anticorpos Monoclonais Humanizados , Ativação do Complemento/imunologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Gangliosídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Síndrome de Miller Fisher/imunologia , Síndrome de Miller Fisher/fisiopatologia , Contração Muscular , Doença Autoimune do Sistema Nervoso Experimental/imunologia , Doença Autoimune do Sistema Nervoso Experimental/fisiopatologia , Junção Neuromuscular/imunologia , Junção Neuromuscular/fisiopatologia , Paralisia Respiratória/imunologia , Paralisia Respiratória/fisiopatologia , Paralisia Respiratória/prevenção & controle , Sinapses/ultraestrutura , Técnicas de Cultura de Tecidos
12.
Br J Haematol ; 142(2): 263-72, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18503589

RESUMO

In paroxysmal nocturnal haemoglobinuria (PNH), chronic destruction of PNH red blood cells (RBCs) by complement leads to anaemia and other serious morbidities. Eculizumab inhibits terminal complement-mediated PNH RBC destruction by targeting C5. In the phase III, double-blind, placebo-controlled, TRIUMPH study, eculizumab reduced haemolysis, stabilized haemoglobin levels, reduced transfusion requirements and improved fatigue in patients with PNH. Herein, we explored the effects of eculizumab on measures of anaemia in patients from the TRIUMPH study and the open-label SHEPHERD study, a more heterogeneous population. Eculizumab reduced haemolysis regardless of pretreatment transfusion requirements and regardless of whether or not patients became transfusion-dependent during treatment (P < 0.001). Reduction in haemolysis was associated with increased PNH RBC counts (P < 0.001) while reticulocyte counts remained elevated. Eculizumab-treated patients demonstrated significantly higher levels of haemoglobin as compared with placebo in TRIUMPH and relative to baseline levels in SHEPHERD (P < 0.001 for each study). Eculizumab lowered transfusion requirement across multiple pretreatment transfusion strata and eliminated transfusion support in a majority of both TRIUMPH and SHEPHERD patients (P < 0.001). Patients who required some transfusion support during treatment with eculizumab showed a reduction in haemolysis and transfusion requirements and an improvement in fatigue. Eculizumab reduces haemolysis and improves anaemia and fatigue, regardless of transfusion requirements.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Hemoglobinúria Paroxística/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais Humanizados , Transfusão de Sangue , Método Duplo-Cego , Contagem de Eritrócitos , Eritrócitos , Fadiga , Feminino , Hemoglobinas/análise , Hemoglobinúria Paroxística/sangue , Hemólise/efeitos dos fármacos , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Adulto Jovem
14.
N Engl J Med ; 350(6): 552-9, 2004 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-14762182

RESUMO

BACKGROUND: Paroxysmal nocturnal hemoglobinuria (PNH) arises from a somatic mutation of the PIG-A gene in a hematopoietic stem cell and the subsequent production of blood cells with a deficiency of surface proteins that protect the cells against attack by the complement system. We tested the clinical efficacy of eculizumab, a humanized antibody that inhibits the activation of terminal complement components, in patients with PNH. METHODS: Eleven transfusion-dependent patients with PNH received infusions of eculizumab (600 mg) every week for four weeks, followed one week later by a 900-mg dose and then by 900 mg every other week through week 12. Clinical and biochemical indicators of hemolysis were measured throughout the trial. RESULTS: Mean lactate dehydrogenase levels decreased from 3111 IU per liter before treatment to 594 IU per liter during treatment (P=0.002). The mean percentage of PNH type III erythrocytes increased from 36.7 percent of the total erythrocyte population to 59.2 percent (P=0.005). The mean and median transfusion rates decreased from 2.1 and 1.8 units per patient per month to 0.6 and 0.0 units per patient per month, respectively (P=0.003 for the comparison of the median rates). Episodes of hemoglobinuria were reduced by 96 percent (P<0.001), and measurements of the quality of life improved significantly. CONCLUSIONS: Eculizumab is safe and well tolerated in patients with PNH. This antibody against terminal complement protein C5 reduces intravascular hemolysis, hemoglobinuria, and the need for transfusion, with an associated improvement in the quality of life in patients with PNH.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Transfusão de Eritrócitos , Hemoglobinúria Paroxística/tratamento farmacológico , Hemólise/efeitos dos fármacos , Adulto , Idoso , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Feminino , Hemoglobinúria/diagnóstico , Hemoglobinúria/tratamento farmacológico , Hemoglobinúria Paroxística/sangue , Hemoglobinúria Paroxística/terapia , Hemoglobinúria Paroxística/urina , Humanos , L-Lactato Desidrogenase/sangue , Masculino , Pessoa de Meia-Idade , Qualidade de Vida , Contagem de Reticulócitos , Urinálise
15.
Shock ; 27(6): 631-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17505302

RESUMO

Many compounds have been shown to prevent reperfusion injury in various animal models, although to date, translation into clinic has revealed several obstacles. Therefore, the National Heart, Lung, and Blood Institute convened a working group to discuss reasons for such failure. As a result, the concept of adequately powered, blinded, randomized studies for preclinical development of a compound has been urged. We investigated the effects of a fibrin-derived peptide Bbeta(15-42) in acute and chronic rodent models of ischemia-reperfusion at three different study centers (Universities of Dusseldorf and Vienna, TNO Biomedical Research). A total of 187 animals were used, and the peptide was compared with the free radical scavenger Tempol, CD18 antibody, alpha-C5 antibody, and the golden standard, ischemic preconditioning. We show that Bbeta(15-42) robustly and reproducibly reduced infarct size in all models of ischemia-reperfusion. Moreover, the peptide significantly reduced plasma levels of the cytokines interleukin 1beta, tumor necrosis factor alpha, and interleukin 6. In rodents, Bbeta(15-42) inhibits proinflammatory cytokine release and is cardioprotective during ischemia-reperfusion injury.


Assuntos
Fibrinogênio/química , Fibrinogênio/fisiologia , Reperfusão Miocárdica , Miocárdio/patologia , Traumatismo por Reperfusão/patologia , Animais , Antígenos CD18/química , Óxidos N-Cíclicos/farmacologia , Modelos Animais de Doenças , Sequestradores de Radicais Livres/química , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Ratos , Marcadores de Spin
17.
Transplantation ; 79(9): 1121-7, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15880054

RESUMO

BACKGROUND: Inhibition of the complement cascade at C5 prevents formation of pro-inflammatory molecules C5a and C5b-9, which play a key role in allograft rejection. The present study was undertaken to determine whether blocking terminal complement with anti-C5 monoclonal antibody (mAb) alone or combined with cyclosporine (CsA) would prevent acute vascular rejection (AVR) in a mouse cardiac allograft model. METHODS: C3H mouse hearts were transplanted into BALB/c mice and randomized into five groups with the following treatments: (1) no treatment; (2) CsA alone; (3) control mAb; (4) anti-C5 mAb alone; and (5) anti-C5 mAb and CsA. RESULTS: Allografts in untreated or control mAb-treated recipients were rapidly rejected at 8.0+/-0.6 and 8.2+/-0.8 days, respectively. These grafts exhibited typical AVR, characterized by vasculitis, hemorrhage, and thrombosis. A high level of complement activity was also demonstrated in these animals. High-dose CsA was not able to inhibit complement activation or AVR, and grafts were rejected in 15.5+/-1.1 days. Anti-C5 monotherapy completely inhibited complement activation and attenuated AVR, but grafts were rejected in 8.3+/-0.5 days by acute cellular rejection. In contrast, a combination of anti-C5 mAb and CsA successfully achieved indefinite graft survival (>100 days). This combined therapy completely inhibited terminal complement activation and prevented both humoral- and cellular-mediated rejection. CONCLUSIONS: Combination therapy of anti-C5 mAb and CsA achieves indefinite graft survival in a mouse cardiac allograft model. These data suggest that inhibition of terminal complement using anti-C5 mAb may be an effective therapeutic adjunct to prevent AVR in clinical transplantation.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complemento C5/imunologia , Ciclosporina/uso terapêutico , Rejeição de Enxerto/prevenção & controle , Transplante de Coração/imunologia , Doença Aguda , Animais , Vasos Sanguíneos/imunologia , Vasos Sanguíneos/patologia , Citometria de Fluxo , Transplante de Coração/patologia , Isoanticorpos/sangue , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Transplante Homólogo/imunologia
18.
Haematologica ; 90(12 Suppl): ECR40, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16464755

RESUMO

Aberrant smooth muscle dystonia during hemolytic episodes in paroxysmal nocturnal hemoglobinuria (PNH) is implicated in the symptoms of abdominal pain, dysphagia and erectile dysfunction. Here we report two PNH patients treated with the complement inhibitor, eculizumab. Complement inhibition has been sustained for over 2 years and results in resolution of intravascular hemolysis and amelioration of symptoms associated with smooth muscle contractions.


Assuntos
Dor Abdominal/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Transtornos de Deglutição/tratamento farmacológico , Disfunção Erétil/tratamento farmacológico , Hemoglobinúria Paroxística/tratamento farmacológico , Músculo Liso/fisiopatologia , Dor Abdominal/etiologia , Adulto , Anemia Aplástica/complicações , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Transfusão de Sangue , Complemento C5/antagonistas & inibidores , Transtornos de Deglutição/etiologia , Disfunção Erétil/etiologia , Fadiga/etiologia , Feminino , Hemoglobinas/efeitos adversos , Hemoglobinúria Paroxística/fisiopatologia , Hemólise , Humanos , Masculino , Pessoa de Meia-Idade , Contração Muscular/efeitos dos fármacos , Óxido Nítrico/fisiologia
19.
JAMA ; 293(13): 1653-62, 2005 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-15811985

RESUMO

CONTEXT: The efficient sequestration of hemoglobin by the red blood cell membrane and the presence of multiple hemoglobin clearance mechanisms suggest a critical need to prevent the buildup of this molecule in the plasma. A growing list of clinical manifestations attributed to hemoglobin release in a variety of acquired and iatrogenic hemolytic disorders suggests that hemolysis and hemoglobinemia should be considered as a novel mechanism of human disease. EVIDENCE ACQUISITION: Pertinent scientific literature databases and references were searched through October 2004 using terms that encompassed various aspects of hemolysis, hemoglobin preparations, clinical symptoms associated with plasma hemoglobin, nitric oxide in hemolysis, anemia, pulmonary hypertension, paroxysmal nocturnal hemoglobinuria, and sickle-cell disease. EVIDENCE SYNTHESIS: Hemoglobin is released into the plasma from the erythrocyte during intravascular hemolysis in hereditary, acquired, and iatrogenic hemolytic conditions. When the capacity of protective hemoglobin-scavenging mechanisms has been saturated, levels of cell-free hemoglobin increase in the plasma, resulting in the consumption of nitric oxide and clinical sequelae. Nitric oxide plays a major role in vascular homeostasis and has been shown to be a critical regulator of basal and stress-mediated smooth muscle relaxation and vasomotor tone, endothelial adhesion molecule expression, and platelet activation and aggregation. Thus, clinical consequences of excessive cell-free plasma hemoglobin levels during intravascular hemolysis or the administration of hemoglobin preparations include dystonias involving the gastrointestinal, cardiovascular, pulmonary, and urogenital systems, as well as clotting disorders. Many of the clinical sequelae of intravascular hemolysis in a prototypic hemolytic disease, paroxysmal nocturnal hemoglobinuria, are readily explained by hemoglobin-mediated nitric oxide scavenging. CONCLUSION: A growing body of evidence supports the existence of a novel mechanism of human disease, namely, hemolysis-associated smooth muscle dystonia, vasculopathy, and endothelial dysfunction.


Assuntos
Doença/etiologia , Hemoglobinas/fisiologia , Hemólise/fisiologia , Óxido Nítrico/fisiologia , Hemoglobinas/farmacologia , Humanos
20.
Blood ; 111(4): 1840-7, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18055865

RESUMO

The terminal complement inhibitor eculizumab was recently shown to be effective and well tolerated in patients with paroxysmal nocturnal hemoglobinuria (PNH). Here, we extended these observations with results from an open-label, non-placebo-controlled, 52-week, phase 3 clinical safety and efficacy study evaluating eculizumab in a broader PNH patient population. Eculizumab was administered by intravenous infusion at 600 mg every 7 +/- 2 days for 4 weeks; 900 mg 7 +/- 2 days later; followed by 900 mg every 14 +/- 2 days for a total treatment period of 52 weeks. Ninety-seven patients at 33 international sites were enrolled. Patients treated with eculizumab responded with an 87% reduction in hemolysis, as measured by lactate dehydrogenase levels (P < .001). Baseline fatigue scores in the FACIT-Fatigue instrument improved by 12.2 +/- 1.1 points (P < .001). Eculizumab treatment led to an improvement in anemia. The increase in hemoglobin level occurred despite a reduction in transfusion requirements from a median of 8.0 units of packed red cells per patient before treatment to 0.0 units per patient during the study (P < .001). Overall, transfusions were reduced 52% from a mean of 12.3 to 5.9 units of packed red cells per patient. Forty-nine patients (51%) achieved transfusion independence for the entire 52-week period. Improvements in hemolysis, fatigue, and transfusion requirements with eculizumab were independent of baseline levels of hemolysis and degree of thrombocytopenia. Quality of life measures were also broadly improved with eculizumab treatment. This study demonstrates that the beneficial effects of eculizumab treatment in patients with PNH are applicable to a broader population of PNH patients than previously studied. This trial is registered at http://clinicaltrials.gov as NCT00130000.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Hemoglobinúria Paroxística/tratamento farmacológico , Adolescente , Adulto , Idoso , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Área Sob a Curva , Transfusão de Sangue , Fadiga/induzido quimicamente , Feminino , Hemoglobinúria Paroxística/sangue , Hemólise/efeitos dos fármacos , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Placebos , Segurança
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa