Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Stroke ; 53(11): 3506-3513, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36128904

RESUMO

It has previously been shown in several animal experiments that platelet GPVI (glycoprotein VI) contributes to thrombosis, particularly in ischemic stroke. Moreover, GPVI levels are upregulated in stroke patients. This review describes the therapeutic roles of anti-GPVI antibody in preclinical models of ischemic stroke and provides the current evidence for potential benefits of glenzocimab, a Fab fragment of humanized anti-GPVI monoclonal antibody, in stroke patients. Anti-GPVI antibody, JAQ1, significantly decreased infarct volume and improved neurological function in mice with transient middle cerebral artery occlusion, a model of ischemic stroke, with no or minor bleeding tendency. Intravenous injection of glenzocimab in nonhuman primates produced rapid inhibition of ex vivo platelet aggregation induced by collagen (a GPVI ligand). Complete platelet inhibition is observed at 30 minutes following administration without increasing the risk of bleeding. In humans, glenzocimab is well tolerated and produces dose-dependent antiplatelet activity. More importantly, glenzocimab (125-1000 mg) was safe when administered as soon as possible (<3 hours) following reperfusion with the r-tPA (recombinant tissue-type plasminogen activator) in patients with acute ischemic stroke. Although glenzocimab 1000 mg (a selected dose) did not demonstrate a significant improvement in overall clinical outcomes, it appeared to provide benefits in severe cases and in patients who required thrombectomy. This promising efficacy together with a good safety profile of glenzocimab warrant further investigation in phase III (ACTISAVE [Adaptive Efficacy and Safety Study of Glenzocimab Used as an Add-On Therapy on Top of Standard of Care in the 4.5 Hours Following an Acute Ischemic Stroke]) clinical study.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Humanos , Animais , Camundongos , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Ativador de Plasminogênio Tecidual/uso terapêutico , Glicoproteínas da Membrana de Plaquetas/uso terapêutico , Ligantes , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Fragmentos Fab das Imunoglobulinas/farmacologia , Fragmentos Fab das Imunoglobulinas/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Colágeno
2.
Thromb Haemost ; 112(1): 26-31, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24553806

RESUMO

Glycoprotein VI (GPVI), a membrane glycoprotein solely expressed in platelets and megakaryocytes, plays a critical role in thrombus formation due to collagen/GPVI-mediated platelet activation and adhesion. Recent studies have shown that surface expression of GPVI on circulating platelets is enhanced in acute cardiovascular diseases such as myocardial infarction and ischaemic stroke. Increased GPVI levels are associated with poor clinical outcome and are an early indicator for imminent myocardial infarction in patients with chest pain. The soluble form of the dimeric GPVI fusion protein (sGPVI-Fc) binds with high affinity to collagen and atherosclerotic plaque tissue. Non-invasive imaging studies with radiolabelled sGPVI-Fc show specific binding activity to vascular lesions in vivo. Further, sGPVI-Fc has been developed as a new therapeutic platelet-based strategy for lesion-directed antithrombotic therapy. This review summarises the potential of GPVI for diagnostic and therapeutic options based on novel non-invasive molecular imaging modalities to ameliorate care of patients with cardiovascular diseases.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Fibrinolíticos/uso terapêutico , Glicoproteínas da Membrana de Plaquetas/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Biomarcadores Farmacológicos/metabolismo , Doenças Cardiovasculares/diagnóstico , Adesão Celular/efeitos dos fármacos , Colágeno/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Modelos Animais , Imagem Molecular , Ativação Plaquetária/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/metabolismo , Medicina de Precisão , Proteínas Recombinantes de Fusão/metabolismo
3.
Circulation ; 125(5): 685-96, 2012 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22223428

RESUMO

BACKGROUND: CXCR4-positive bone marrow cells (BMCs) are critically involved in cardiac repair mechanisms contributing to preserved cardiac function. Stromal cell-derived factor-1 (SDF-1) is the most prominent BMC homing factor known to augment BMC engraftment, which is a limiting step of stem cell-based therapy. After myocardial infarction, SDF-1 expression is rapidly upregulated and promotes myocardial repair. METHODS AND RESULTS: We have established a bifunctional protein consisting of an SDF-1 domain and a glycoprotein VI (GPVI) domain with high binding affinity to the SDF-1 receptor CXCR4 and extracellular matrix proteins that become exposed after tissue injury. SDF1-GPVI triggers chemotaxis of CXCR4-positive cells, preserves cell survival, enhances endothelial differentiation of BMCs in vitro, and reveals proangiogenic effects in ovo. In a mouse model of myocardial infarction, administration of the bifunctional protein leads to enhanced recruitment of BMCs, increases capillary density, reduces infarct size, and preserves cardiac function. CONCLUSIONS: These results indicate that administration of SDF1-GPVI may be a promising strategy to treat myocardial infarction to promote myocardial repair and to preserve cardiac function.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Quimiocina CXCL12/farmacologia , Coração/efeitos dos fármacos , Coração/fisiopatologia , Ataque Isquêmico Transitório/terapia , Infarto do Miocárdio/terapia , Glicoproteínas da Membrana de Plaquetas/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/uso terapêutico , Colágeno/metabolismo , Ataque Isquêmico Transitório/patologia , Ataque Isquêmico Transitório/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/uso terapêutico , Ligação Proteica , Receptores CXCR4/metabolismo , Resultado do Tratamento
5.
Basic Res Cardiol ; 103(4): 356-67, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18431526

RESUMO

Platelet adhesion to the atherosclerotic vascular wall induces thrombosis and boosters vascular inflammation and atheroprogression. In the present study we studied the binding of the platelet collagen receptor glycoprotein (GP) VI to human atherosclerotic plaques (AP) and the role of GPVI-mediated platelet adhesion for atheroprogression. Soluble GPVI-Fc fusion protein bound to immobilized collagen type I, collagen type III, and predominantly to the core region of human carotid atheromatous plaques. The pattern of GPVI-Fc binding was similar to the immunostaining pattern of collagen type III and differed from the immunostaining of collagen type I, which was more intense in the cap than in the core. Plaque-induced platelet aggregation in stirred blood and platelet adhesion/aggregate formation under flow were inhibited by the anti-GPVI monoclonal antibody 5C4 or by pretreatment of plaques with anti-collagen type I and anti-collagen type III antibody, or GPVI-Fc. However, there was no correlation between GPVI-Fc binding and platelet aggregating activity of individual plaques. GPVI bound also to atherosclerotic arteries of ApoE-deficient mice in vivo as assessed by small animal positron emission tomography (PET). Prolonged administration of soluble GPVI attenuated atheroprogression in ApoE-deficient mice. In humans, GPVI binding to collagenous type I and type III structures of the plaque core region mediates plaque-induced platelet adhesion and aggregation, but GPVI binding is not the sole platelet-activating determinant of plaques. In mice, GPVI-mediated platelet adhesion to the atherosclerotic vascular wall is involved in atheroprogression in vivo. Taken together, our data suggests that GPVI is a relevant target to prevent atherothrombotic events and atheroprogression.


Assuntos
Aterosclerose/etiologia , Colágeno/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Animais , Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Adesividade Plaquetária , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/uso terapêutico
6.
Blood ; 105(4): 1492-9, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15507524

RESUMO

Glycoprotein VI (GPVI) is an essential platelet collagen receptor; therefore, the inhibition of GPVI-collagen interactions may be an attractive antithrombotic strategy. We have previously shown that targeting of GPVI with antibodies leads to the depletion of the receptor and to long-term antithrombotic protection in mice. An alternative agent to interfere with GPVI-collagen interactions might be soluble GPVI acting as a competitive inhibitor, thereby averting undesired effects on platelets. To test this, we expressed soluble dimeric human GPVI, comprising the extracellular domain of the receptor fused to the human immunoglobulin Fc domain (GPVI-Fc), and compared its antithrombotic potential with that of anti-GPVI antibodies in mice. In contrast to a recent report, we found by intravital fluorescence microscopy and ultrasonic flow measurements that GPVI-Fc had no effect on platelet adhesion and thrombus formation at the injured arterial wall, whereas anti-GPVI antibodies profoundly inhibited these processes. Similar results were obtained with a fusion protein comprising the extracellular domain of mouse GPVI and human IgG-Fc. This indicates that direct targeting of GPVI provides significantly stronger protection against arterial thrombosis than soluble GPVI dimer.


Assuntos
Fibrinolíticos/farmacologia , Isoanticorpos/farmacologia , Glicoproteínas da Membrana de Plaquetas/imunologia , Glicoproteínas da Membrana de Plaquetas/farmacologia , Animais , Sítios de Ligação de Anticorpos , Ligação Competitiva/fisiologia , Lesões das Artérias Carótidas/sangue , Lesões das Artérias Carótidas/imunologia , Lesões das Artérias Carótidas/terapia , Colágeno/metabolismo , Colágeno/farmacologia , Dimerização , Fibrinolíticos/metabolismo , Fibrinolíticos/uso terapêutico , Humanos , Fragmentos Fc das Imunoglobulinas/metabolismo , Injeções Intravenosas , Isoanticorpos/metabolismo , Isoanticorpos/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Adesividade Plaquetária/fisiologia , Agregação Plaquetária/fisiologia , Inibidores da Agregação Plaquetária/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Glicoproteínas da Membrana de Plaquetas/genética , Glicoproteínas da Membrana de Plaquetas/uso terapêutico , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Solubilidade , Trombose/sangue , Trombose/imunologia , Trombose/prevenção & controle
7.
Crit Care Med ; 28(3): 638-42, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10752807

RESUMO

OBJECTIVE: To evaluate the safety and efficacy of the platelet-activating factor receptor antagonist BB-882 in the treatment of patients with sepsis. DESIGN: Double-blind, placebo-controlled, randomized, multi-centered study. SETTING: Thirty-four European intensive care units. PATIENTS: One hundred fifty-two patients with clinical suspicion of infection and a mean APACHE II score between 15 and 35 in the 24 hrs before entry into the trial. INTERVENTIONS: Patients received either a loading dose of 4 mg of BB-882 on the first day, followed by an intravenous infusion of 96 mg/24 hrs for up to 120 hrs, or placebo. MEASUREMENTS: Hemodynamic, respiratory and oxygen transport variables, blood lactate concentrations, interleukin-6, interleukin-8, tumor necrosis factor (TNF)-alpha, soluble TNF receptor concentrations, organ failure score, 28-day mortality rate, Acute Physiology And Chronic Health Evaluation (APACHE) II score within 24 hrs of entry. RESULTS: Sixty-nine patients (42 male, 27 female) received placebo and 83 (59 male, 24 female) received BB-882. Patients ranged in age from 16 to 89 yrs (mean, 60 yrs). No important differences existed between the two groups in terms of gender distribution, age, or initial APACHE II score. Sepsis was identified as Gram-positive in 49 patients, Gram-negative in 40, mixed in 37, and unknown in 26. No important differences were shown in hemodynamic, respiratory, or oxygen transport variables between groups during the study. Organ failure scores were similar in the two groups throughout the study. Cytokine concentrations were not significantly different in the two groups. Within 28 days of entering the study, 75 patients died, including 31 (45%) in the placebo group and 44 (53%) in the treatment group, p = .32. The median time to death in the placebo group was 6.0 days, and in the treatment group, it was 4.5 days (p = .30). CONCLUSION: Treatment of sepsis with the platelet-activating factor antagonist BB-882 offers no advantage over placebo on survival, hemodynamic status, respiratory function, or organ failure scores.


Assuntos
Leucina/análogos & derivados , Fator de Ativação de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/uso terapêutico , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Sepse/tratamento farmacológico , APACHE , Adulto , Idoso , Idoso de 80 Anos ou mais , Citocinas/sangue , Método Duplo-Cego , Europa (Continente)/epidemiologia , Feminino , Hemodinâmica , Humanos , Imunoterapia/métodos , Infusões Intravenosas , Ácido Láctico/sangue , Leucina/uso terapêutico , Masculino , Pessoa de Meia-Idade , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Testes de Função Respiratória , Sepse/mortalidade , Análise de Sobrevida
8.
Eur J Pharmacol ; 376(3): 293-300, 1999 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-10448890

RESUMO

In cases of severe human scorpion envenoming, lung injury is a common finding and frequently the cause of death. In the rat, two distinct mechanisms account for oedema following the intravenous injection of the venom -- acute left ventricular failure resulting from a massive release of catecholamines and an increase in pulmonary vascular permeability. In the present work, we investigated the effects of a tachykinin NK1 receptor antagonist (CP96,345, the dihydrochloride salt of (2S,3S)-cis-2-(diphenylmethyl)-N-((2-methoxyphenyl)methyl)-1-az abicycol[2.2.2]octan-3-amine) and its 2 R-3 R inactive enantiomer (CP96,344) on the acute lung injury induced by the i.v. injection of Tityus serrulatus venom in rats. Lung injury was assessed by evaluating the extravasation of Evans blue dye in the bronchoalveolar lavage fluid and in the lung of venom-treated and control animals. The effects of the platelet-activating factor (PAF) receptor antagonist WEB2170 (2-methyl-1-phenylimidazol[4,5c]pyridine) were evaluated for comparison. The i.v. injection of the venom induced the extravasation of Evans blue in the bronchoalveolar lavage fluid and into the left lung. Pretreament with the tachykinin NK1 receptor antagonist CP96,345, but not CP96,344, inhibited Evans blue dye extravasation in the bronchoalveolar lavage fluid and in the lung by 96% and 86%, respectively. The PAF receptor antagonist WEB2170 inhibited the increase in vascular permeability in the bronchoalveolar lavage fluid by 60% and had no effect on the extravasation to the lung parenchyma of venom-injected animals. In addition to abrogating lung injury, pretreatment of rats with CP96,345, but not CP96,344 or WEB2170, decreased by 70% the mortality induced by the venom. This is the first study to show the relevance of the tachykinin NK1 receptor in mediating lung injury and mortality in animals injected with the neurotoxic T. serrulatus venom. Blockade of the tachykinin NK1 receptor may represent an important strategy in the treatment of patients with signs of severe envenoming and clearly deserves further studies.


Assuntos
Azepinas/uso terapêutico , Compostos de Bifenilo/uso terapêutico , Glicoproteínas da Membrana de Plaquetas/uso terapêutico , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Receptores de Taquicininas/uso terapêutico , Síndrome do Desconforto Respiratório/tratamento farmacológico , Venenos de Escorpião , Triazóis/uso terapêutico , Animais , Cobaias , Íleo/efeitos dos fármacos , Masculino , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Ratos , Ratos Wistar , Receptores de Taquicininas/antagonistas & inibidores , Síndrome do Desconforto Respiratório/induzido quimicamente
9.
Prog Cardiovasc Dis ; 40(2): 141-58, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9327830

RESUMO

A large number of drug trials for prevention of restenosis have been conducted with many showing little or conflicting benefit. Antiplatelets such as aspirin, ticlopidine and thromboxane A2 receptor inhibitors have not shown a clear benefit. Similarly, antithrombotics, either acting indirectly such as heparin, or as direct thrombin inhibitors such as hirudin and hirulog, do not prevent restenosis. Trials with ACE inhibitors, HMG-CoA reductase inhibitors and fish-oil supplements have yielded inconclusive results. The antiproliferatives, angiopeptin, trapidil and tranilast have shown some benefit in small-scale studies. Other drug classes of potential benefit include the glycoprotein IIb/IIIa receptor antagonists, inhibitors of the early coagulation cascade, calcium channel blockers and nitric oxide donors. Drug research into restenosis prevention has been hampered by problems with the definition of restenosis and the applicability in humans of animal models. Although no single drug has conclusively proven effective yet, the promise of a number of agents, together with other nonpharmacological strategies will likely result in further reductions in the incidence of restenosis.


Assuntos
Angioplastia Coronária com Balão/efeitos adversos , Doença das Coronárias/terapia , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/patologia , Abciximab , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Anticorpos Monoclonais/uso terapêutico , Anticoagulantes/uso terapêutico , Fármacos Cardiovasculares/uso terapêutico , Constrição Patológica , Angiografia Coronária , Doença das Coronárias/prevenção & controle , Modelos Animais de Doenças , Humanos , Hipolipemiantes/uso terapêutico , Fragmentos Fab das Imunoglobulinas/uso terapêutico , Oligopeptídeos/uso terapêutico , Peptídeos Cíclicos , Inibidores da Agregação Plaquetária/uso terapêutico , Glicoproteínas da Membrana de Plaquetas/uso terapêutico , Recidiva , Somatostatina/análogos & derivados , Somatostatina/uso terapêutico , Trapidil/uso terapêutico
10.
Eur J Gastroenterol Hepatol ; 8(6): 569-73, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8823572

RESUMO

OBJECTIVE: To assess the anti-inflammatory action of lexipafant (BB-882), a platelet activating factor antagonist, in an animal model of acute colitis. DESIGN: An animal intervention study. METHODS: Following the rectal instillation of formalin 0.75% into male New Zealand White (NZW) rabbits, 0.85 ml of aggregated immunoglobulin was administered i.v. Treatment groups (0.8 mg/kg, n = 6; 2.4 mg/kg, n = 13; 3.2 mg/kg, n = 10) were given bolus doses of BB-882 two-hourly i.v. (control group, n = 25). Rectal dialysis was performed before induction of colitis and sacrifice. Dialysate leukotriene B4 (LTB4), prostaglandin E2 (PGE2) and thromboxane B2 (TXB2) levels were determined. Tissue was saved for histology and measurement of myeloperoxidase content. RESULTS: There was a dose-dependent improvement in macroscopic scores (2.4 and 3.2 mg/kg: P < 0.02, P < 0.001) and myeloperoxidase levels (3.2 mg/kg: P < 0.04). Dialysate LTB4 levels fell (2.4 and 3.2 mg/kg: P < 0.03, P < 0.02) as did PGE2 levels. TXB2 concentrations remained unaffected. CONCLUSION: The PAF receptor antagonist BB-882 shows efficacy in treating inflammation in an animal model of acute colitis as evidenced by a dose-dependent fall in macroscopic mucosal damage, neutrophil infiltration and reduced generation of inflammatory mediators.


Assuntos
Colite/tratamento farmacológico , Mucosa Intestinal/efeitos dos fármacos , Leucina/análogos & derivados , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Animais , Colite/induzido quimicamente , Dinoprostona/análise , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Formaldeído , Injeções Intravenosas , Mucosa Intestinal/patologia , Leucina/administração & dosagem , Leucina/uso terapêutico , Leucotrieno B4/análise , Masculino , Glicoproteínas da Membrana de Plaquetas/uso terapêutico , Coelhos , Tromboxano B2/análise
11.
Surgery ; 117(4): 458-65, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7536354

RESUMO

BACKGROUND: Neutrophils have been shown to play a role in ischemia-reperfusion injury, and the initial interaction of neutrophils with the endothelium is mediated through the selectin family of adhesion molecules. Thus the purpose of these studies was to determine whether a P-selectin-IgG chimera was protective in a model of ischemia-reperfusion injury. METHODS: The model used was a rabbit ear model of ischemia-reperfusion. Selectin-IgG chimeras were given at the time of reperfusion of the tissue, and their efficacy was compared with an anti-CD18 antibody (MHM23). RESULTS: The P-selectin-IgG was as protective in this model as an anti-CD18 antibody. The chimera did not mediate its effect by causing the animals to become neutropenic. CONCLUSIONS: P-selectin plays a role in ischemia-reperfusion injury. This is in agreement with data from other groups. The fact that the chimera was effective in this model suggests that carbohydrates or small molecule mimics of carbohydrates would be effective in this model. Such antiinflammatory agents may have fewer side effects in terms of increased risk of sepsis.


Assuntos
Imunoglobulina G/uso terapêutico , Isquemia/fisiopatologia , Glicoproteínas da Membrana de Plaquetas/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Animais , Antígenos CD18/imunologia , Moléculas de Adesão Celular/uso terapêutico , Orelha , Endotélio Vascular/fisiopatologia , Isquemia/patologia , Masculino , Neutrófilos/fisiologia , Selectina-P , Coelhos , Traumatismo por Reperfusão/patologia
12.
Adv Pediatr Infect Dis ; 9: 37-58, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-8123225

RESUMO

Attempts to augment immune function in infants or children with overwhelming sepsis or immune deficiency have focused on recombinant cytokines and interferons. Although early in their clinical use, the proinflammatory and antiinflammatory effects of the interleukins show promise in regulating the acute inflammatory response. The hematopoietic growth factors have demonstrated considerable clinical effect, especially in individuals with distinct hematopoietic disorders and in patients receiving immunosuppressive chemotherapy. The interferons gamma and alpha have received considerable attention over the last decade as potential immunomodulators. The stimulatory effects of INF-gamma on human neutrophils suggest its therapeutic usefulness in children with specific neutrophil dysfunction, and INF-alpha has shown broad clinical application both as an antitumor as well as an antiviral agent. The most recent studies of the integrins and selectins implicate their potential role in regulating a number of infectious disease states. As we learn more about the complexity of interactions and the delicate balance these molecules have in regulating the immune response, we will be better able to implement their use in regulating disease states in infants and children.


Assuntos
Citocinas/uso terapêutico , Infecções/tratamento farmacológico , Infecções Oportunistas Relacionadas com a AIDS/prevenção & controle , Criança , Pré-Escolar , Fatores Estimuladores de Colônias/uso terapêutico , Humanos , Lactente , Infecções/imunologia , Integrinas , Interferon-alfa/uso terapêutico , Interferon gama/uso terapêutico , Glicoproteínas da Membrana de Plaquetas/uso terapêutico
13.
Am J Cardiol ; 72(15): 1121-5, 1993 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-8237799

RESUMO

Hemorrhagic events remain the most worrisome complication in patients receiving drugs that alter hemostasis for treatment of acute coronary syndromes. The 7E3 Fab monoclonal antibody provides a dose-dependent inhibition of platelet aggregation via the glycoprotein IIb/IIIa receptor. This study examines the correlation of hemostatic parameters with bleeding events in patients receiving intravenous 7E3 while enrolled in acute myocardial infarction and high-risk percutaneous transluminal coronary angioplasty pilot studies. Patients with acute myocardial infarction received 100 mg of tissue-type plasminogen activator over 3 hours followed by an escalating intravenous bolus dose of murine 7E3 (0.1 mg/kg [n = 5], 0.2 mg/kg [n = 22], 0.15 mg/kg [n = 13], 0.25 mg/kg [n = 20]). Patients in the high-risk angioplasty trial received a chimeric 7E3 bolus (up to 0.25 mg/kg) with (n = 32) or without (n = 15) intravenous continuous infusion of 7E3 (10 micrograms/min for 6 to 24 hours) after elective angioplasty. Patients in both studies received aspirin therapy (325 mg/day) and partial thromboplastin time-guided (1.5 to 2 times normal) heparin infusion. Bleeding events occurred in 34 of 124 patients (27%). The median template bleeding times (minutes) for patients in the groups with bleeding versus no bleeding events in the trials was 13.5 versus 14 and 30 versus 30, respectively (p = NS). In patients with myocardial infarction, a substantial decline in platelet count at 24 hours was associated with bleeding (p = 0.02).(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Tempo de Sangramento , Hemorragia/prevenção & controle , Glicoproteínas da Membrana de Plaquetas/imunologia , Adolescente , Adulto , Idoso , Animais , Feminino , Hemorragia/sangue , Hemorragia/etiologia , Humanos , Modelos Lineares , Masculino , Camundongos , Pessoa de Meia-Idade , Projetos Piloto , Glicoproteínas da Membrana de Plaquetas/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/uso terapêutico , Valor Preditivo dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...