Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 141(10): 1147-1158, 2023 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-36108308

RESUMEN

Factor VIII (FVIII) circulates in a noncovalent complex with von Willebrand Factor (VWF), the latter determining FVIII half-life. The VWF-binding aptamer rondaptivon pegol (BT200) increases plasma levels of VWF/FVIII in healthy volunteers. This trial assessed its safety, pharmacokinetics, and pharmacodynamics in hemophilia A. Nineteen adult patients (ages 20-62 years, 4 women) with hemophilia A (8 mild, 2 moderate, and 9 severe) received subcutaneous injections of rondaptivon pegol. After an initial fixed dose of 3 mg on days 0 and 4, patients received weekly doses of 2 to 9 mg until day 28. Severe hemophilia A patients underwent sparse-sampling population pharmacokinetics individual profiling after the final dose of rondaptivon pegol. Adverse events, pharmacokinetics, and pharmacodynamics were assessed. FVIII activity and VWF levels were measured. All patients tolerated rondaptivon pegol well. The geometric mean half-life of rondaptivon pegol was 5.4 days and rondaptivon pegol significantly increased VWF levels. In severe hemophilia A, 6 doses of rondaptivon pegol increased the half-lives of 5 different FVIII products from a median of 10.4 hours to 31.1 hours (range, 20.8-56.0 hours). Median FVIII increased from 22% to 48% in mild hemophilia A and from 3% to 7.5% in moderate hemophilia A. Rondaptivon pegol is a first-in-class prohemostatic molecule that extended the half-life of substituted FVIII approximately 3-fold and increased endogenous FVIII levels approximately 2-fold in hemophilia patients. This trial was registered at www.clinicaltrials.gov as #NCT04677803.


Asunto(s)
Hemofilia A , Hemostáticos , Adulto , Humanos , Femenino , Adulto Joven , Persona de Mediana Edad , Factor de von Willebrand/uso terapéutico , Hemofilia A/tratamiento farmacológico , Factor VIII , Hemostáticos/uso terapéutico , Semivida
2.
Haematologica ; 107(9): 2121-2132, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34818873

RESUMEN

Von Willebrand factor (VWF) and factor VIII (FVIII) circulate in a noncovalent complex in blood and promote primary hemostasis and clotting, respectively. A new VWF A1-domain binding aptamer, BT200, demonstrated good subcutaneous bioavailability and a long half-life in non-human primates. This first-in-human, randomized, placebo-controlled, doubleblind trial tested the hypothesis that BT200 is well tolerated and has favorable pharmacokinetic and pharmacodynamic effects in 112 volunteers. Participants received one of the following: a single ascending dose of BT200 (0.18-48 mg) subcutaneously, an intravenous dose, BT200 with concomitant desmopressin or multiple doses. Pharmacokinetics were characterized, and the pharmacodynamic effects were measured by VWF levels, FVIII clotting activity, ristocetin-induced aggregation, platelet function under high shear rates, and thrombin generation. The mean half-lives ranged from 7-12 days and subcutaneous bioavailability increased dose-dependently exceeding 55% for doses of 6-48 mg. By blocking free A1 domains, BT200 dose-dependently decreased ristocetin-induced aggregation, and prolonged collagen-adenosine diphosphate and shear-induced platelet plug formation times. However, BT200 also increased VWF antigen and FVIII levels 4-fold (P<0.001), without increasing VWF propeptide levels, indicating decreased VWF/FVIII clearance. This, in turn, increased thrombin generation and accelerated clotting. Desmopressin-induced VWF/FVIII release had additive effects on a background of BT200. Tolerability and safety were generally good, but exaggerated pharmacology was seen at saturating doses. This trial identified a novel mechanism of action for BT200: BT200 dose-dependently increases VWF/FVIII by prolonging half-life at doses well below those which inhibit VWF-mediated platelet function. This novel property can be exploited therapeutically to enhance hemostasis in congenital bleeding disorders.


Asunto(s)
Enfermedades de von Willebrand , Factor de von Willebrand , Desamino Arginina Vasopresina , Factor VIII , Humanos , Ristocetina/farmacología , Trombina , Factor de von Willebrand/metabolismo
3.
Blood ; 133(9): 893-901, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30559259

RESUMEN

Cold agglutinin disease is a difficult-to-treat autoimmune hemolytic anemia in which immunoglobulin M antibodies bind to erythrocytes and fix complement, resulting in predominantly extravascular hemolysis. This trial tested the hypothesis that the anti-C1s antibody sutimlimab would ameliorate hemolytic anemia. Ten patients with cold agglutinin disease participated in the phase 1b component of a first-in-human trial. Patients received a test dose of 10-mg/kg sutimlimab followed by a full dose of 60 mg/kg 1 to 4 days later and 3 additional weekly doses of 60 mg/kg. All infusions were well tolerated without premedication. No drug-related serious adverse events were observed. Seven of 10 patients with cold agglutinin disease responded with a hemoglobin increase >2 g/dL. Sutimlimab rapidly increased hemoglobin levels by a median of 1.6 g/dL within the first week, and by a median of 3.9 g/dL (interquartile range, 1.3-4.5 g/dL; 95% confidence interval, 2.1-4.5) within 6 weeks (P = .005). Sutimlimab rapidly abrogated extravascular hemolysis, normalizing bilirubin levels within 24 hours in most patients and normalizing haptoglobin levels in 4 patients within 1 week. Hemolytic anemia recurred when drug levels were cleared from the circulation 3 to 4 weeks after the last dose of sutimlimab. Reexposure to sutimlimab in a named patient program recapitulated the control of hemolytic anemia. All 6 previously transfused patients became transfusion-free during treatment. Sutimlimab was safe, well tolerated, and rapidly stopped C1s complement-mediated hemolysis in patients with cold agglutinin disease, significantly increasing hemoglobin levels and precluding the need for transfusions. This trial was registered at www.clinicaltrials.gov as #NCT02502903.


Asunto(s)
Anemia Hemolítica Autoinmune/tratamiento farmacológico , Anemia Hemolítica/prevención & control , Anticuerpos Monoclonales Humanizados/uso terapéutico , Complemento C1s/antagonistas & inhibidores , Hemólisis/efectos de los fármacos , Índice de Severidad de la Enfermedad , Anciano , Anemia Hemolítica/etiología , Anemia Hemolítica Autoinmune/complicaciones , Complemento C1s/inmunología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos
4.
Blood Adv ; 6(18): 5467-5476, 2022 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-35772170

RESUMEN

Type 2B von Willebrand disease (VWD) is characterized by an increased binding affinity of von Willebrand factor (VWF) to platelet glycoprotein Ib. This can lead to clearance of high-molecular-weight (HMW) multimers and thrombocytopenia with a resulting moderate-severe bleeding phenotype. Rondoraptivon pegol (BT200) is a pegylated aptamer binding to the A1 domain of VWF with a novel mechanism of action: it enhances VWF/factor VIII (FVIII) levels by decreasing their clearance. To study the potential benefit of rondoraptivon pegol in patients with type 2B VWD, we conducted a prospective phase 2 trial. Patients with type 2B VWD received 3 mg rondoraptivon pegol subcutaneously on study days 1, 4, and 7, followed by 6 to 9 mg every week until day 28. Five patients (male:female ratio = 3:2) were included. Rondoraptivon pegol rapidly tripled platelet counts from a median of 60 to 179 × 10E9/L (P < .001). Circulating VWF antigen increased from a median of 64% to 143%, which doubled FVIII activity levels from 67% to 134%. In all thrombocytopenic patients, plasma levels of VWF:GPIbM normalized, VWF ristocetin cofactor and VWF collagen-binding activity increased, and HMW multimers appeared. These pronounced improvements reversed during washout of the drug, thus demonstrating causality. The A1 domain binding aptamer directly corrects the underlying defect of type 2B VWD, thus providing a novel potential option for prophylaxis and treatment of patients with this VWD type. These data provide the basis for a phase 2b/3 trial in such patients. This trial was registered at www.clinicaltrials.gov as #NCT04677803.


Asunto(s)
Hemostáticos , Enfermedad de von Willebrand Tipo 2 , Enfermedades de von Willebrand , Colágeno , Factor VIII/uso terapéutico , Femenino , Hemostáticos/uso terapéutico , Humanos , Masculino , Recuento de Plaquetas , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Polietilenglicoles/uso terapéutico , Estudios Prospectivos , Enfermedades de von Willebrand/tratamiento farmacológico , Factor de von Willebrand/metabolismo
5.
Am J Hematol ; 86(1): 87-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20981675

RESUMEN

Despite improvements in our understanding of the pathophysiology of thrombotic thrombocytopenic purpura (TTP), little data exist regarding the long-term sequelae following a diagnosis of TTP. We present the results of a comprehensive evaluation of neurologic injury that included a magnetic resonance imaging (MRI), a neurocognitive testing, and an evaluation of health-related quality of life. Twenty-seven patients with a history of idiopathic TTP functioning normally in their activities of daily living were recruited from existing patient cohorts at both the Ohio State University (n 5 12) (Columbus) and the University College London Hospitals (n 5 15) (London, UK). Nine of 23 (39%) of the MRI studies were abnormal; 17/27 (63%) patients demonstrated neurocognitive impairment, particularly in tests of visual learning and memory. Health-related quality of life scores were also significantly lower than age- and gender-matched US norms for both the composite mental component score and physical component score. These data suggest that the prevalence of neurologic findings in TTP patients in remission is quite high and is largely undetected by routine clinical evaluations. Further longitudinal study will be required to define the risk for neurologic injury and the long-term prognosis in patients previously diagnosed with TTP.


Asunto(s)
Enfermedades del Sistema Nervioso/etiología , Púrpura Trombocitopénica Trombótica/complicaciones , Adolescente , Adulto , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Enfermedades del Sistema Nervioso/diagnóstico , Pronóstico , Púrpura Trombocitopénica Trombótica/fisiopatología , Calidad de Vida , Encuestas y Cuestionarios , Adulto Joven
6.
Sci Rep ; 11(1): 3092, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33542410

RESUMEN

The effect of conventional anti-platelet agents is limited in secondary stroke prevention, and their effects are blunted under high shear stress in the presence of increased levels of circulating von Willebrand factor (VWF). VWF is critically involved in thrombus formation at sites of stenotic extracranial/intracranial arteries. A third generation anti-VWF aptamer (BT200) has been generated which could be useful for secondary stroke prevention. To characterize the effects of BT200 in blood of patients with large artery atherosclerosis stroke (LAA). Blood samples were obtained from 33 patients with acute stroke or transient ischemic attack to measure inhibition of VWF activity and VWF-dependent platelet function. Patients who received clopidogrel or dual antiplatelet therapy did not differ in VWF dependent platelet function tests from aspirin treated patients. Of 18 patients receiving clopidogrel with or without aspirin, only 3 had a prolonged collagen adenosine diphosphate closure time, and none of the patients had ristocetin induced aggregation in the target range. BT200 concentration-dependently reduced median VWF activity from 178 to < 3%, ristocetin induced platelet aggregation from 40U to < 10U and prolonged collagen adenosine diphosphate closure times from 93 s to > 300 s. Baseline VWF activity correlated (r = 0.86, p < 0.001) with concentrations needed to reduce VWF activity to < 20% of normal, indicating that BT200 acts in a target concentration-dependent manner. Together with a long half-life supporting once weekly administration, the safety and tolerability observed in an ongoing phase I trial, and the existence of a reversal agent, BT200 is an interesting drug candidate.


Asunto(s)
Aptámeros de Péptidos/farmacología , Ataque Isquémico Transitorio/sangre , Accidente Cerebrovascular/tratamiento farmacológico , Factor de von Willebrand/efectos de los fármacos , Anciano , Aspirina/uso terapéutico , Plaquetas/efectos de los fármacos , Colágeno/metabolismo , Femenino , Humanos , Arteriosclerosis Intracraneal/sangre , Arteriosclerosis Intracraneal/complicaciones , Arteriosclerosis Intracraneal/patología , Ataque Isquémico Transitorio/patología , Ataque Isquémico Transitorio/prevención & control , Masculino , Agregación Plaquetaria/efectos de los fármacos , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/patología , Trombosis/sangre , Trombosis/tratamiento farmacológico , Trombosis/etiología , Trombosis/patología
7.
Transfusion ; 50(5): 1079-87, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20070617

RESUMEN

BACKGROUND: In thrombotic thrombocytopenic purpura (TTP), ultralarge von Willebrand factor (VWF) multimers bind platelet (PLT) glycoprotein Ib and lead to the formation of disseminated fibrin-poor, VWF-rich PLT thrombi. The aptamer ARC1779 blocks binding of the VWF A1 domain to PLT glycoprotein Ib. We evaluated whether ARC1779 inhibits the excessive VWF activity and VWF-mediated PLT function in patients with TTP. STUDY DESIGN AND METHODS: We studied the ex vivo concentration response curves for ARC1779 on PLT function analyzer (PFA-100, Dade Behring) and cone-and-plate analyzer (CPA, Impact-R) PLT function tests, agonist-induced PLT aggregation, and VWF activity of TTP patients (n = 11, three in acute phase and eight in remission) and healthy controls (n = 44). RESULTS: VWF activity and VWF-dependent PLT plug formation were increased in TTP patients relative to healthy controls, but agonist-induced PLT aggregation was not. ARC1779 blocked collagen/adenosine 5'-diphosphate (ADP)-induced PLT plug formation as measured by PFA-100 with an inhibitory concentration (IC)(100) of approximately 1 microg/mL in citrate-anticoagulated samples and approximately 3 to 4 microg/mL in hirudin-anticoagulated samples. A similar concentration of ARC1779 was necessary to block shear-dependent PLT adhesion in both TTP patients and healthy controls using the CPA assay (IC(100) of approx. 1 microg/mL for both). ARC1779 blocked VWF activity with an IC(90) of approximately 3 to 4 microg/mL in all subjects, but did not inhibit PLT aggregation by ADP, collagen, or arachidonic acid even at concentrations much greater than those that fully inhibited VWF-dependent PLT function. CONCLUSIONS: ARC1779 potently and specifically inhibits VWF activity and VWF-dependent PLT function. ARC1779 may be a promising novel therapeutic for the treatment of TTP.


Asunto(s)
Plaquetas/efectos de los fármacos , Púrpura Trombocitopénica Trombótica/tratamiento farmacológico , Factor de von Willebrand/antagonistas & inhibidores , Adulto , Aptámeros de Nucleótidos , Plaquetas/fisiología , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Agregación Plaquetaria/efectos de los fármacos , Púrpura Trombocitopénica Trombótica/sangre
8.
J Thromb Haemost ; 18(7): 1695-1704, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32275107

RESUMEN

BACKGROUND: BT200, a pegylated form of the aptamer BT100, inhibits binding of von Willebrand factor (VWF) to platelet glycoprotein GPIb, preventing arterial thrombosis in cynomolgus monkeys. It is being developed for secondary prevention of arterial thrombosis such as stroke or myocardial infarction. Inhibition of thrombogenesis by BT200 is expected to provide a therapeutic benefit. However, there may be unexpected bleeding (eg, incidental trauma) in which a reversal agent is required. To address this need, BT101, a complementary aptamer, has been developed to specifically inhibit BT100 and BT200 function. OBJECTIVES: To characterize the effects of BT101 both in vitro and in vivo. METHODS: The direct interaction between BT101 and the core aptamer BT100 was evaluated using polyacrylamide gel electrophoresis. The binding of BT200 to purified human VWF and inhibition of VWF activity was further characterized using enzyme-linked immunosorbent assay. VWF-dependent platelet function was measured by the platelet function analyzer and aggregometry in whole blood. In addition, both the in vivo pharmacokinetic profile of BT101 as well as its ability to reverse BT200 activity, were evaluated in cynomolgus monkeys. RESULTS: BT101 bound to the core aptamer BT100 at a 1:1 ratio, inhibited BT200 binding to purified human VWF, and reversed BT200-induced inhibition of both VWF activity and VWF-dependent platelet function in vitro. After intravenous injection to monkeys, BT101 reversed BT200-induced effects on VWF activity and platelet function within minutes, without causing any adverse effects. CONCLUSIONS: The results of this study demonstrate that BT101 is an effective reversal agent for BT200.


Asunto(s)
Trombosis , Factor de von Willebrand , Animales , Plaquetas , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria , Trombosis/tratamiento farmacológico
9.
Sci Rep ; 10(1): 11180, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32636459

RESUMEN

Von Willebrand factor (VWF) plays a major role in arterial thrombosis. Antiplatelet drugs induce only a moderate relative risk reduction after atherothrombosis, and their inhibitory effects are compromised under high shear rates when VWF levels are increased. Therefore, we investigated the ex vivo effects of a third-generation anti-VWF aptamer (BT200) before/after stimulated VWF release. We studied the concentration-effect curves BT200 had on VWF activity, platelet plug formation under high shear rates (PFA), and ristocetin-induced platelet aggregation (Multiplate) before and after desmopressin or endotoxin infusions in healthy volunteers. VWF levels increased > 2.5-fold after desmopressin or endotoxin infusion (p < 0.001) and both agents elevated circulating VWF activity. At baseline, 0.51 µg/ml BT200 reduced VWF activity to 20% of normal, but 2.5-fold higher BT200 levels were required after desmopressin administration (p < 0.001). Similarly, twofold higher BT200 concentrations were needed after endotoxin infusion compared to baseline (p < 0.011). BT200 levels of 0.49 µg/ml prolonged collagen-ADP closure times to > 300 s at baseline, whereas 1.35 µg/ml BT200 were needed 2 h after desmopressin infusion. Similarly, twofold higher BT200 concentrations were necessary to inhibit ristocetin induced aggregation after desmopressin infusion compared to baseline (p < 0.001). Both stimuli elevated plasma VWF levels in a manner representative of thrombotic or pro-inflammatory conditions such as arterial thrombosis. Even under these conditions, BT200 potently inhibited VWF activity and VWF-dependent platelet function, but higher BT200 concentrations were required for comparable effects relative to the unstimulated state.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Plaquetas/efectos de los fármacos , Agregación Plaquetaria , Factor de von Willebrand/antagonistas & inhibidores , Adenosina Difosfato/metabolismo , Adulto , Plaquetas/metabolismo , Plaquetas/fisiología , Células Cultivadas , Colágeno/metabolismo , Desamino Arginina Vasopresina/farmacología , Femenino , Humanos , Lipopolisacáridos/farmacología , Masculino , Persona de Mediana Edad , Factor de von Willebrand/metabolismo
10.
J Thromb Haemost ; 18(5): 1113-1123, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32011054

RESUMEN

BACKGROUND: Thrombus formation involves coagulation proteins and platelets. The latter, referred to as platelet-mediated thrombogenesis, is predominant in arterial circulation. Platelet thrombogenesis follows vascular injury when extracellular von Willebrand factor (VWF) binds via its A3 domain to exposed collagen, and the free VWF A1 domain binds to platelet glycoprotein Ib (GPIb). OBJECTIVES: To characterize the antiplatelet/antithrombotic activity of the pegylated VWF antagonist aptamer BT200 and identify the aptamer VWF binding site. METHODS: BT100 is an optimized aptamer synthesized by solid-phase chemistry and pegylated (BT200) by standard conjugation chemistry. The affinity of BT200 for purified human VWF was evaluated as was VWF inhibition in monkey and human plasma. Efficacy of BT200 was assessed in the monkey FeCl3 femoral artery thrombosis model. RESULTS: BT200 bound human VWF at an EC50 of 5.0 nmol/L and inhibited VWF A1 domain activity in monkey and human plasma with mean IC50 values of 183 and 70 nmol/L. BT200 administration to cynomolgus monkeys caused a time-dependent and dose-dependent effect on VWF A1 domain activity and inhibited platelet function as measured by collagen adenosine diphosphate closure time in the platelet function analyzer. BT200 demonstrated a bioavailability of ≥77% and exhibited a half-life of >100 hours after subcutaneous injection. The treatment effectively prevented arterial occlusion in an FeCl3 -induced thrombosis model in monkeys. CONCLUSIONS: BT200 has shown promising inhibition of human VWF in vitro and prevented arterial occlusion in non-human primates. These data including a long half-life after subcutaneous injections provide a strong rationale for ongoing clinical development of BT200.


Asunto(s)
Trombosis , Factor de von Willebrand , Animales , Plaquetas , Humanos , Pruebas de Función Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria , Trombosis/tratamiento farmacológico
11.
Thromb Haemost ; 120(9): 1282-1290, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32679592

RESUMEN

BACKGROUND: von Willebrand factor (VWF) is crucial for arterial thrombosis and its plasma levels are increased in acute coronary syndromes (ACSs). The effects of conventional platelet inhibitors are compromised by elevated VWF under high shear rates. BT200 is a third-generation aptamer that binds and inhibits the A1 domain of human VWF. This article aims to study whether VWF is a predictor of mortality in ACS patients under potent P2Y12 blocker therapy and to examine the effects of a VWF inhibiting aptamer BT200 and its concentrations required to inhibit VWF in plasma samples of patients with ACS. METHODS: VWF activity was measured in 320 patients with ACS, and concentration effect curves of BT200 were established in plasma pools containing different VWF concentrations. RESULTS: Median VWF activity in patients was 170% (interquartile range % confidence interval [CI]: 85-255) and 44% of patients had elevated (> 180%) VWF activity. Plasma levels of VWF activity predicted 1-year (hazard ratio [HR]: 2.68; 95% CI: 1.14-6.31; p < 0.024) and long-term (HR: 2.59; 95% CI: 1.10-6.09) mortality despite treatment with potent platelet inhibitors (dual-antiplatelet therapy with aspirin and prasugrel or ticagrelor). Although half-maximal concentrations were 0.1 to 0.2 µg/mL irrespective of baseline VWF levels, increasing concentrations (0.42-2.13 µg/mL) of BT200 were needed to lower VWF activity to < 20% of normal in plasma pools containing increasing VWF activity (p < 0.001). CONCLUSION: VWF is a predictor of all-cause mortality in ACS patients under contemporary potent P2Y12 inhibitor therapy. BT200 effectively inhibited VWF activity in a target concentration-dependent manner.


Asunto(s)
Síndrome Coronario Agudo/tratamiento farmacológico , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico , Factor de von Willebrand/antagonistas & inhibidores , Factor de von Willebrand/metabolismo , Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/metabolismo , Síndrome Coronario Agudo/mortalidad , Anciano , Aptámeros de Nucleótidos/farmacología , Biomarcadores/sangre , Biomarcadores/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inhibidores de Agregación Plaquetaria/farmacología , Receptores Purinérgicos P2Y12/metabolismo , Factor de von Willebrand/análisis
12.
Blood Adv ; 4(6): 997-1005, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32176765

RESUMEN

Cold agglutinin disease (CAD) causes predominantly extravascular hemolysis and anemia via complement activation. Sutimlimab is a novel humanized monoclonal antibody directed against classical pathway complement factor C1s. We aimed to evaluate the safety and efficacy of long-term maintenance treatment with sutimlimab in patients with CAD. Seven CAD patients treated with sutimlimab as part of a phase 1B study were transitioned to a named patient program. After a loading dose, patients received biweekly (once every 2 weeks) infusions of sutimlimab at various doses. When a patient's laboratory data showed signs of breakthrough hemolysis, the dose of sutimlimab was increased. Three patients started with a dose of 45 mg/kg, another 3 with 60 mg/kg, and 1 with a fixed dose of 5.5 g every other week. All CAD patients responded to re-treatment, and sutimlimab increased hemoglobin from a median initial level of 7.7 g/dL to a median peak of 12.5 g/dL (P = .016). Patients maintained near normal hemoglobin levels except for a few breakthrough events that were related to underdosing and which resolved after the appropriate dose increase. Four of the patients included were eventually treated with a biweekly 5.5 g fixed-dose regimen of sutimlimab. None of them had any breakthrough hemolysis. All patients remained transfusion free while receiving sutimlimab. There were no treatment-related serious adverse events. Overlapping treatment with erythropoietin, rituximab, or ibrutinib in individual patients was safe and did not cause untoward drug interactions. Long-term maintenance treatment with sutimlimab was safe, effectively inhibited hemolysis, and significantly increased hemoglobin levels in re-exposed, previously transfusion-dependent CAD patients.


Asunto(s)
Anemia Hemolítica Autoinmune , Complemento C1s , Anemia Hemolítica Autoinmune/tratamiento farmacológico , Activación de Complemento , Hemólisis , Humanos , Rituximab
13.
Circulation ; 117(11): 1449-59, 2008 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-18347221

RESUMEN

von Willebrand factor (VWF) plays a pivotal role in platelet adhesion and aggregation at sites of high shear rates (eg, in coronary arteries that have stenotic or ruptured atherosclerotic plaque lesions). Numerous studies have investigated the relationship between VWF plasma levels and thromboembolic cardiovascular events. In contrast to the rather weak association in the general population, in patients with preexisting vascular disease, VWF is significantly predictive for adverse cardiac events, including death. Likewise, VWF typically rises during the course of acute coronary syndrome, and the extent of this VWF release is an independent predictor of adverse clinical outcome in these patients. Various lines of evidence indicate that VWF is not only a marker but also actually an important effector in the pathogenesis of myocardial infarction. This central role of VWF in thrombogenesis has made it a promising target for research into new antiplatelet therapies that specifically inhibit VWF. This review focuses on the role of VWF in acute coronary syndrome and further outlines the relevance of therapeutic interventions targeting VWF for acute coronary syndrome patients.


Asunto(s)
Enfermedades Cardiovasculares/sangre , Trombosis/sangre , Factor de von Willebrand/fisiología , Enfermedad Aguda , Angioplastia Coronaria con Balón , Biomarcadores , Enfermedades Cardiovasculares/epidemiología , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/complicaciones , Trombosis Coronaria/sangre , Trombosis Coronaria/etiología , Endotelio Vascular/lesiones , Endotelio Vascular/metabolismo , Hemorreología , Humanos , Infarto del Miocardio/sangre , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/terapia , Activación Plaquetaria/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Receptores de Superficie Celular/antagonistas & inhibidores , Factores de Riesgo , Rotura Espontánea , Síndrome , Terapia Trombolítica , Factor de von Willebrand/análisis
14.
Transfusion ; 49(10): 2181-5, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19538431

RESUMEN

BACKGROUND: Plasma exchange is the main therapy for thrombotic thrombocytopenic purpura (TTP). No treatments other than plasma exchange have been documented to be effective nor are approved for treatment of TTP. The anti-von Willebrand factor (VWF) aptamer ARC1779 effectively inhibits VWF activity in plasma samples of TTP patients and thus shear-dependent platelet (PLT) function as measured by the PLT function analyzer PFA-100 (Dade Behring). It was hypothesized that ARC1779 would offer a potentially effective treatment option for a critically ill patient, refractory to standard care. CASE REPORT: A 39-year-old male patient with idiopathic TTP, refractory to daily plasma exchange, rituximab, steroids, and splenectomy, was additionally treated with a continuous infusion of the anti-von Willebrand factor (VWF) aptamer ARC1779 for 3 weeks. RESULTS: Plasma concentrations of approximately 10 microg/mL ARC1779 decreased VWF activity by more than 96%. Plasma exchange treatment acutely decreased the plasma concentrations of ARC1779 by a mean of 47% (range, 40%-61%). Thus, additional minibolus infusions of ARC1779 were given after each plasma exchange to rapidly restore steady-state concentrations. ARC1779 resulted in an increase of PLT counts as long as ARC1779 was given. On three occasions the infusion was stopped, each accompanied by a decrease in PLT counts and worsening of microangiopathy. No serious adverse effects were observed during the treatment with ARC1779. CONCLUSION: ARC1779 caused a clear and reproducible increase in PLT counts in an otherwise refractory TTP case. These clinical, pharmacokinetic, and pharmacodynamic data provide a rational basis for clinical trials with ARC1779 in TTP.


Asunto(s)
Aptámeros de Nucleótidos/uso terapéutico , Fibrinolíticos/uso terapéutico , Púrpura Trombocitopénica Trombótica/tratamiento farmacológico , Factor de von Willebrand/antagonistas & inhibidores , Adulto , Humanos , Masculino , Púrpura Trombocitopénica Trombótica/sangre , Púrpura Trombocitopénica Trombótica/patología
15.
Platelets ; 20(5): 334-40, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19637097

RESUMEN

ARC1779 is an aptamer, which blocks binding of the von Willebrand Factor (VWF) A1 domain to platelet GPIb receptors. VWF is increased in the elderly an in the setting of acute myocardial infarction (AMI), as reflected by increased shear-dependent platelet function. We hypothesized that ARC1779 concentration-dependently inhibits ex vivo platelet function, and that this concentration effect relationship may be shifted in patients with AMI. We studied ex vivo dose response curves for ARC1779 on VWF activity, shear-dependent platelet function, and agonist-induced platelet aggregation. We included patients with AMI on standard treatment (n = 40), young (n = 20) and elderly controls (n = 20) in this ex vivo dosing study. AMI patients displayed approximately 2-fold increased plasma levels of VWF activity as compared to controls. ARC1779 inhibited VWF activity (IC90: approximately 3-4 microg/mL) and shear dependent platelet function (Platelet Function Analyzer (PFA-100), IC50: approximately 0.5-0.9 microg/mL and Cone and Plate Analyzer (CPA), IC50: approximately 0.1-0.4 microg/mL in citrated blood) at comparable concentrations in all groups. In contrast to GPIIb/IIIa antagonists, ARC1779 did not inhibit platelet aggregation induced by ADP, collagen or arachidonic acid at concentrations (10 microg/mL) that fully inhibited VWF dependent platelet function. ARC1779 potently and specifically inhibits VWF activity and VWF dependent platelet function, even in the setting of AMI where VWF activity is increased.


Asunto(s)
Aptámeros de Nucleótidos/uso terapéutico , Infarto del Miocardio/sangre , Agregación Plaquetaria/efectos de los fármacos , Factor de von Willebrand/antagonistas & inhibidores , Adulto , Factores de Edad , Anciano , Aptámeros de Nucleótidos/farmacología , Células Sanguíneas , Plaquetas/fisiología , Estudios de Casos y Controles , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/tratamiento farmacológico , Adulto Joven
16.
J Invest Dermatol ; 139(12): 2417-2424.e2, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31229501

RESUMEN

Deposition of autoantibodies (α-BP180 and BP230) and complement along the dermal-epidermal-junction is a hallmark of bullous pemphigoid and was shown to be important for pathogenesis. Given the adverse effects of standard treatment (glucocorticoids, immunosuppressants), there is an unmet need for safe and effective therapies. In this phase 1 trial, we evaluated the safety and activity of BIVV009 (sutimlimab, previously TNT009), a targeted C1s inhibitor, in 10 subjects with active or past bullous pemphigoid (NCT02502903). Four weekly 60 mg/kg infusions of BIVV009 proved sufficient for inhibition of the classical complement pathway in all patients, as measured by CH50. C3c deposition along the dermal-epidermal junction was partially or completely abrogated in 4 of 5 patients, where it was present at baseline. BIVV009 was found to be safe and tolerable in this elderly population, with only mild to moderate adverse events reported (e.g., headache, fatigue). One serious adverse event (i.e., fatal cardiac decompensation) occurred at the end of the post-treatment observation period in an 84-year-old patient with a history of diabetes and heart failure, but was deemed unlikely to be related to the study drug. This trial provides the first results with a complement-targeting therapy in bullous pemphigoid, to our knowledge, and supports further studies on BIVV009's efficacy and safety in this population.


Asunto(s)
Autoantígenos/administración & dosificación , Complemento C3/metabolismo , Vía Clásica del Complemento/efectos de los fármacos , Dermis/patología , Distonina/administración & dosificación , Epidermis/patología , Colágenos no Fibrilares/administración & dosificación , Penfigoide Ampolloso/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Dermis/metabolismo , Epidermis/metabolismo , Femenino , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Penfigoide Ampolloso/metabolismo , Penfigoide Ampolloso/patología , Colágeno Tipo XVII
17.
Circulation ; 116(23): 2678-86, 2007 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-18025536

RESUMEN

BACKGROUND: ARC1779 is a therapeutic aptamer antagonist of the A1 domain of von Willebrand Factor (vWF), the ligand for receptor glycoprotein 1b on platelets. ARC1779 is being developed as a novel antithrombotic agent for use in patients with acute coronary syndromes. METHODS AND RESULTS: This was a randomized, double-blind, placebo-controlled study in 47 healthy volunteers of doses of ARC1779 from 0.05 to 1.0 mg/kg. Pharmacodynamic effects were measured by an ELISA for free vWF A1 binding sites and by a platelet function analyzer. In terms of pharmacokinetics, the concentration-time profile of ARC1779 appeared monophasic. The observed concentration and area under the curve were dose proportional. The mean apparent elimination half-life was approximately 2 hours, and mean residence time was approximately 3 hours. The mean apparent volumes of distribution (at steady state and during terminal phase) were approximately one half the blood volume, suggesting that ARC1779 distribution is in the central compartment. The mean clearance ranged from approximately 10% to approximately 21% of the glomerular filtration rate, suggesting that renal filtration may not be a major mechanism of clearance of ARC1779. Inhibition of vWF A1 binding activity was achieved with an EC(90) value of 2.0 mug/mL (151 nmol/L) and of platelet function with an EC(90) value of 2.6 mug/mL (196 nmol/L). ARC1779 was generally well tolerated, and no bleeding was observed. Adverse events tended to be minor and not dose related. CONCLUSIONS: This is the first-in-human evaluation of a novel aptamer antagonist of vWF. ARC1779 produced dose- and concentration-dependent inhibition of vWF activity and platelet function with duration of effect suitable for the intended clinical use in acute coronary syndromes.


Asunto(s)
Síndrome Coronario Agudo/tratamiento farmacológico , Aptámeros de Nucleótidos/farmacocinética , Fibrinolíticos/farmacocinética , Factor de von Willebrand/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Aptámeros de Nucleótidos/efectos adversos , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Fibrinolíticos/efectos adversos , Humanos , Masculino , Persona de Mediana Edad , Pruebas de Función Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/agonistas , Estructura Terciaria de Proteína , Factores de Tiempo
18.
Adv Drug Deliv Rev ; 134: 36-50, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30321620

RESUMEN

Aptamers are synthetic molecules structured as single-stranded DNA or RNA oligonucleotides that can be designed to mimic the functional properties of monoclonal antibodies. They bind to the target molecules (typically soluble or cell-bound proteins) with high affinity (with picomolar to low nanomolar range) and specificity, and therefore can be an alternative to therapeutic antibodies or peptide ligands. This paper reviews published data regarding pharmacokinetics, pharmacodynamics and safety of aptamers from preclinical and clinical studies. Aptamers have been developed for the treatment of a variety of diseases, including cancer, macular degeneration,g cardiovascular disease, diabetes and anaemia of chronic diseases. There are several preclinical studies with unmodified aptamers, but the vast majority of aptamer trials in humans have been conducted with modified aptamers, because unmodified aptamers demonstrate metabolic instability, as well as rapid renal filtration and elimination. Various strategies have been developed to improve the pharmacokinetic profile of aptamers. Aside from chemical modification of nucleotides in order to stabilize them against nuclease degradation, the main modification to extend the half-life is pegylation. Therefore, the process of pegylation as well as its benefits and possible shortcomings will briefly be discussed.


Asunto(s)
Aptámeros de Nucleótidos/efectos adversos , Aptámeros de Nucleótidos/farmacocinética , Neoplasias/tratamiento farmacológico , Anemia de Células Falciformes/tratamiento farmacológico , Aptámeros de Nucleótidos/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Diabetes Mellitus/tratamiento farmacológico , Humanos
19.
Thromb Haemost ; 118(6): 959-978, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29847840

RESUMEN

Despite great efforts in stroke research, disability and recurrence rates in ischaemic stroke remain unacceptably high. To address this issue, one potential target for novel therapeutics is the glycoprotein von Willebrand factor (vWF), which increases in thrombogenicity especially under high shear rates as it bridges between vascular sub-endothelial collagen and platelets. The rationale for vWF as a potential target in stroke comes from four bodies of evidence. (1) Animal models which recapitulate the pathogenesis of stroke and validate the concept of targeting vWF for stroke prevention and the use of the vWF cleavage enzyme ADAMTS13 in acute stroke treatment. (2) Extensive epidemiologic data establishing the prognostic role of vWF in the clinical setting showing that high vWF levels are associated with an increased risk of first stroke, stroke recurrence or stroke-associated mortality. As such, vWF levels may be a suitable marker for further risk stratification to potentially fine-tune current risk prediction models which are mainly based on clinical and imaging data. (3) Genetic studies showing an association between vWF levels and stroke risk on genomic levels. Finally, (4) studies of patients with primary disorders of excess or deficiency of function in the vWF axis (e.g. thrombotic thrombocytopenic purpura and von Willebrand disease, respectively) which demonstrate the crucial role of vWF in atherothrombosis. Therapeutic inhibition of VWF by novel agents appears particularly promising for secondary prevention of stroke recurrence in specific sub-groups of patients such as those suffering from large artery atherosclerosis, as designated according to the TOAST classification.


Asunto(s)
Isquemia/tratamiento farmacológico , Accidente Cerebrovascular/tratamiento farmacológico , Factor de von Willebrand/uso terapéutico , Proteína ADAMTS13/uso terapéutico , Animales , Modelos Animales de Enfermedad , Medicina Basada en la Evidencia , Predisposición Genética a la Enfermedad , Humanos , Isquemia/genética , Prevención Secundaria , Accidente Cerebrovascular/genética
20.
Clin Pharmacol Ther ; 104(4): 655-663, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29737533

RESUMEN

Aberrant activation of the classical complement pathway is the common underlying pathophysiology of orphan diseases such as bullous pemphigoid, antibody-mediated rejection of organ transplants, cold agglutinin disease, and warm autoimmune hemolytic anemia. Therapeutic options for these complement-mediated disorders are limited and sutimlimab, a humanized monoclonal antibody directed against complement factor C1s, may be potentially useful for inhibition of the classical complement pathway. A phase I, first-in-human, double-blind, randomized, placebo-controlled, dose-escalation trial of single and multiple doses of sutimlimab or placebo was conducted in 64 volunteers to evaluate safety, tolerability, pharmacokinetic, and pharmacodynamic profiles. Single and multiple infusions of sutimlimab were well tolerated without any safety concerns. sutimlimab exhibited a steep concentration-effect relationship with a Hill coefficient of 2.4, and an IC90 of 15.5 µg/mL. This study establishes the foundation for using sutimlimab as a highly selective inhibitor of the classical complement pathway in different diseases.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Complemento C1s/antagonistas & inhibidores , Inactivadores del Complemento/administración & dosificación , Vía Clásica del Complemento/efectos de los fármacos , Adulto , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Austria , Complemento C1s/inmunología , Inactivadores del Complemento/efectos adversos , Inactivadores del Complemento/farmacocinética , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Esquema de Medicación , Femenino , Voluntarios Sanos , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA