Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
PLoS One ; 18(1): e0269738, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36607961

RESUMEN

INTRODUCTION: Abnormal coagulation and inflammation are hallmarks of SARs-COV-19. Stratifying affected patients on admission to hospital may help identify those who at are risk of developing severe disease early on. Rotational Thromboelastometry (ROTEM) is a point of care test that can be used to measure abnormal coagulation and calprotectin is a measure of inflammation. AIM: Assess if ROTEM can measure hypercoagulability on admission and identify those who will develop severe disease early on. Assess if calprotectin can measure inflammation and if there is a correlation with ROTEM and calprotectin. METHODS: COVID-19 patients were recruited on admission and ROTEM testing was undertaken daily for a period of 7 days. Additionally inflammatory marker calprotectin was also tested for the same period. RESULTS: 33 patients were recruited to the study out of which 13 were admitted to ITU and 20 were treated on the ward. ROTEM detected a hypercoagulable state on admission but did not stratify between those admitted to a ward or escalated to ITU. Calprotectin levels were raised but there was no statistical difference (p = 0.73) between patients admitted to a ward or escalated to ITU. Significant correlations were observed between FIBA5 (r = 0.62; p<0.00), FIBCFT (r = -0.57; p<0.00), FIBMCF (r = 0.64; p<0.00) and INMCF (r = 0.57; p<0.00) and calprotectin. CONCLUSION: COVID-19 patients were hypercoagulable on admission. The correlations between ROTEM and calprotectin underline the interactions between inflammation and coagulation.


Asunto(s)
COVID-19 , Trombofilia , Humanos , Tromboelastografía , COVID-19/complicaciones , COVID-19/diagnóstico , Proyectos Piloto , Trombofilia/diagnóstico , Inflamación
4.
J Neurotrauma ; 38(8): 1177-1184, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30526281

RESUMEN

We aim to describe the outcomes after chronic subdural hematoma drainage (CSDH) management in a large cohort of patients on antithrombotic drugs, either antiplatelets or anticoagulants, at presentation and to inform clinical decision making on the timing of surgery and recommencement of these drugs. We used data from a previous UK-based multi-center, prospective cohort study. Outcomes included recurrence within 60 days, functional outcome at discharge, and thromboembolic event during hospital stay. We performed Cox regression on recurrence and multiple logistic regression on functional outcome. There were 817 patients included in the analysis, of which 353 (43.2%) were on an antithrombotic drug at presentation. We observed a gradual reduction in risk of recurrence for patients during the 6 weeks post-CSDH surgery. Neither antiplatelet nor anticoagulant drug use influenced risk of CSDH recurrence (hazard ratio, 0.93; 95% confidence interval [CI], 0.58-1.48; p = 0.76) or persistent/worse functional impairment (odds ratio, 1.08; 95% CI, 0.76-1.55; p = 0.66). Delaying surgery after cessation of antiplatelet drug did not affect risk of bleed recurrence. There were 15 in-hospital thromboembolic events recorded. Events were more common in the group pre-treated with antithrombotic drugs (3.3%) compared to the non-antithrombotic group (0.9%). Patients on an antithrombotic drug pre-operatively were at higher risk of thromboembolic events with no excess risk of bleed recurrence or worse functional outcome after CSDH drainage. The data did not support delaying surgery in patients on antithrombotic therapy. In the absence of a randomized controlled trial, early surgery and early antithrombotic recommencement should be considered in those at high risk of thromboembolic events.


Asunto(s)
Anticoagulantes/administración & dosificación , Drenaje/tendencias , Hematoma Subdural Crónico/tratamiento farmacológico , Hematoma Subdural Crónico/cirugía , Inhibidores de Agregación Plaquetaria/administración & dosificación , Anciano , Anciano de 80 o más Años , Anticoagulantes/efectos adversos , Estudios de Cohortes , Drenaje/efectos adversos , Femenino , Hematoma Subdural Crónico/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , Inhibidores de Agregación Plaquetaria/efectos adversos , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/prevención & control , Cuidados Preoperatorios/efectos adversos , Cuidados Preoperatorios/métodos , Estudios Prospectivos , Factores de Riesgo , Resultado del Tratamiento , Trepanación/efectos adversos , Trepanación/métodos
6.
Blood ; 136(11): 1347-1350, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32746455

RESUMEN

The association of severe coronavirus disease 2019 (COVID-19) with an increased risk of venous thromboembolism (VTE) has resulted in specific guidelines for its prevention and management. The VTE risk appears highest in those with critical care admission. The need for postdischarge thromboprophylaxis remains controversial, which is reflected in conflicting expert guideline recommendations. Our local protocol provides thromboprophylaxis to COVID-19 patients during admission only. We report postdischarge VTE data from an ongoing quality improvement program incorporating root-cause analysis of hospital-associated VTE (HA-VTE). Following 1877 hospital discharges associated with COVID-19, 9 episodes of HA-VTE were diagnosed within 42 days, giving a postdischarge rate of 4.8 per 1000 discharges. Over 2019, following 18 159 discharges associated with a medical admission; there were 56 episodes of HA-VTE within 42 days (3.1 per 1000 discharges). The odds ratio for postdischarge HA-VTE associated with COVID-19 compared with 2019 was 1.6 (95% confidence interval, 0.77-3.1). COVID-19 hospitalization does not appear to increase the risk of postdischarge HA-VTE compared with hospitalization with other acute medical illness. Given that the risk-benefit ratio of postdischarge thromboprophylaxis remains uncertain, randomized controlled trials to evaluate the role of continuing thromboprophylaxis in COVID-19 patients following hospital discharge are required.


Asunto(s)
Betacoronavirus/aislamiento & purificación , Infecciones por Coronavirus/complicaciones , Hospitalización/estadística & datos numéricos , Alta del Paciente/estadística & datos numéricos , Neumonía Viral/complicaciones , Tromboembolia Venosa/etiología , COVID-19 , Infecciones por Coronavirus/virología , Estudios de Seguimiento , Humanos , Pandemias , Neumonía Viral/virología , Pronóstico , SARS-CoV-2 , Tromboembolia Venosa/patología
7.
Blood Coagul Fibrinolysis ; 28(4): 295-302, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27607599

RESUMEN

: Cardiac surgery induces a multifactorial coagulopathy. Regular use of tranexamic acid (TXA) is becoming standard of care. Clinical challenges include selecting optimal dosing regimen and balancing the benefit versus risk of additional dosing with antifibrinolytics. The objective was to evaluate the effect of TXA by assessing kinetic properties of plasma clot formation, clot stability, and clot fibrinolysis. The study was a prospective case follow-up of 28 patients undergoing cardiac surgery (mean age 63.9 years, 29% women). Blood samples were analysed at seven time points during the first 48 h after surgery. All patients were treated with TXA, 2 g at start surgery, 1 g during extra corporeal circulation, and 1 g after reversal of heparin. An automated clot lysis assay using tissue factor and tissue plasminogen activator (tPA) was performed to evaluate clot formation, stability, and fibrinolysis. TXA protects against facilitated fibrinolysis and induces up to 13-fold increase in clot stability. All patients showed complete resistance to tPA-induced fibrinolysis during the first 6 h after cardiac surgery declining to 33% of patients at 48 h. Impaired renal function was associated with prolonged resistance to tPA-induced fibrinolysis. Despite inhibition of fibrinolysis with TXA, the overall clot stability declines and the kinetic properties of clot formation were impaired after cardiac surgery. TXA induces a multifold increase in clot resistance to fibrinolysis but does not affect clot formation or clot stability. Monitoring the level of resistance to fibrinolysis may prevent overdosing in particular in patients with impaired renal function.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Procedimientos Quirúrgicos Cardíacos/métodos , Tiempo de Lisis del Coágulo de Fibrina , Fibrinólisis/efectos de los fármacos , Ácido Tranexámico/administración & dosificación , Anciano , Monitoreo de Drogas/métodos , Femenino , Humanos , Cuidados Intraoperatorios/métodos , Cinética , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Factores de Tiempo , Ácido Tranexámico/farmacología
8.
Ann Hematol ; 93(4): 683-92, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24193375

RESUMEN

Haemophilia is characterised by defective thrombin generation, reduced clot stability and spontaneous bleeding. Treatment with factor VIII (FVIII) concentrate or bypassing agents (e.g. recombinant factor VIIa (rFVIIa)) is generally effective. Occasionally, haemostasis is not achieved, which may reflect a failure of factor concentrate to normalise clot stability. Tranexamic acid (TXA) is often used to aid haemostasis in surgery (e.g. joint replacements and dental procedures). Used routinely as an adjunct, it may enhance clot stability and allow effective, reliable, and cost-effective treatment at lower doses of factor concentrate. This study hypothesised that clot stabilising adjunct TXA is required in addition to factor substitution to normalise clot stability in whole blood from patients with severe haemophilia A. The in vitro effect of varying concentrations of recombinant FVIII or recombinant FVIIa and adjunct TXA on whole blood clot stability was measured by thromboelastometry. Coagulation was triggered by tissue factor and clots were challenged with tissue plasminogen activator. The area under the elasticity curve was the primary endpoint. High concentrations of FVIII and rFVIIa increased clot stability to levels that were not significantly different from controls (Mean ± SD: control 112,694 ± 84,115; FVIII 78,662 ± 74,126; rFVIIa 95,918 ± 88,492). However, the response was highly variable between individuals and demonstrates why some patients show clinical resistance to treatment. Addition of TXA resulted in normalised clot stability in all individuals, even when combined with the lowest doses of factor concentrate. The results support the concept that a more efficient, reliable and cost effective treatment may be obtained if TXA is combined with factor concentrates to treat individuals with haemophilia.


Asunto(s)
Antifibrinolíticos/administración & dosificación , Factor VIII/administración & dosificación , Factor VIIa/administración & dosificación , Hemofilia A/tratamiento farmacológico , Ácido Tranexámico/administración & dosificación , Adolescente , Adulto , Quimioterapia Combinada , Hemofilia A/sangre , Hemofilia A/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/administración & dosificación , Ácido Tranexámico/sangre , Resultado del Tratamiento , Adulto Joven
9.
Thromb Res ; 131(5): e210-3, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23473642

RESUMEN

INTRODUCTION: Fibrinogen deficiency often develops during massive bleeding due to e.g. fluid resuscitation with colloid plasma expanders like hydroxyethyl starch. This study investigates the haemostatic effect of various sources of fibrinogen: fibrinogen concentrates (Haemocomplettan® (FC 1), CSL Behring and Clottagen® (FC 2), LFB Biomedicaments), fresh frozen plasma (FFP), and Cryoprecipitate (CP). METHODS: Whole blood samples provided by healthy individuals (n=8) were diluted 1:1 with HES or 0.9% isotonic saline as control. Various sources of fibrinogen was added to the HES diluted samples in concentrations equivalent to giving a 70 kg male either no correction, 2 grams of FC 1 or FC 2, 4 grams of FC 1 or FC 2, 15 ml/kg of FFP, 2 packs of CP or 4 packs of CP. Haemostatic effect was assessed by thromboelastometry initiated by tissue factor, with maximum clot firmness (MCF) as the primary endpoint. In addition thrombin generation was assessed for each intervention RESULTS: HES dilution reduced MCF significantly more than isotonic saline. High dose FC 1, 2 and CP corrected the MCF so that it did not differ significantly from the isotonic saline dilution group. Thrombin generation following isotonic saline and HES dilution was comparable and not decreased compared to whole blood. Fibrinogen concentrates supplementation did not increase thrombin generation whereas CP and FFP both increased thrombin generation CONCLUSION: Fibrinogen concentrates investigated dose dependently and equally corrected the MCF and caused no increase in thrombin generation. Cryoprecipitate also corrected the MCF, but also increased thrombin generation. FFP failed to improve MCF, but increased thrombin generation.


Asunto(s)
Afibrinogenemia/terapia , Fibrinógeno/administración & dosificación , Hemodilución/métodos , Plasma , Hemodilución/efectos adversos , Humanos , Masculino , Tromboelastografía/métodos , Trombina/metabolismo
10.
Semin Thromb Hemost ; 38(3): 268-73, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22460909

RESUMEN

Coagulation factor I (fibrinogen) plays an essential role in the hemostatic system by bridging activated platelets and being the key substrate for thrombin in establishing a consolidating fibrin network. Fibrinogen is synthesized in the liver and the plasma concentration is 1 to 5-4.0 g/L. During recent 10 years, fibrinogen has been recognized to play an important role in controlling hemorrhage. Dilutional coagulopathy induced by colloid plasma expanders is characterized by fibrinogen deficiency and dysfunctional fibrin polymerization. Trauma and use of extracorporeal circulation is also known to reduce levels of fibrinogen. A series of laboratory experiments and experimental animal studies have suggested fibrinogen as a potent hemostatic agent. These data are supported by retrospective surveys as well as a series of prospective proof of principal clinical trials. This article provides a description of the biochemistry and mechanisms of fibrinogen as well as the etiology for developing fibrinogen deficiency. Furthermore, it summarizes laboratory and experimental data on the role of fibrinogen in dilutional coagulopathy and addresses laboratory monitoring issues. Finally, it lists retrospective and prospective studies, which have been designed to assess the clinical efficacy and safety of hemostatic intervention with fibrinogen concentrate.


Asunto(s)
Fibrinógeno/uso terapéutico , Hemostáticos/uso terapéutico , Humanos , Estudios Prospectivos , Estudios Retrospectivos
11.
Haematologica ; 97(10): 1570-3, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22419578

RESUMEN

The Philadelphia negative myeloproliferative neoplasms, including polycythemia vera, essential thrombocythemia, and myelofibrosis, are associated with substantial vascular and transformative complications. Standard therapy for high-risk disease, particularly in patients that have failed initial therapy, remains controversial. Non-pegylated interferon has previously been shown to be effective in controlling erythrocytosis, thrombocytosis and thrombotic complications, but was found to have poor tolerability and excessive adverse effects. Recently, pegylated interferon alpha-2a was introduced and found to be better tolerated and less toxic than standard interferon. In addition, in recent phase II trials, pegylated interferon alpha-2a therapy was found to induce both hematologic and molecular remissions. We retrospectively analyzed 118 myeloproliferative patients who underwent pegylated interferon alpha-2a treatment. Responses were evaluated by ELN, IWG-MET and EUMNET standardized criteria sets and adverse effects were analyzed.


Asunto(s)
Antineoplásicos/uso terapéutico , Interferón-alfa/uso terapéutico , Trastornos Mieloproliferativos/tratamiento farmacológico , Polietilenglicoles/uso terapéutico , Adolescente , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Estudios de Cohortes , Femenino , Humanos , Interferón-alfa/administración & dosificación , Interferón-alfa/efectos adversos , Masculino , Persona de Mediana Edad , Trastornos Mieloproliferativos/patología , Estadificación de Neoplasias , Polietilenglicoles/administración & dosificación , Polietilenglicoles/efectos adversos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Resultado del Tratamiento , Adulto Joven
12.
Blood ; 119(15): 3622-8, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22234684

RESUMEN

Solulin is a soluble form of thrombomodulin that is resistant to proteolysis and oxidation. It has been shown to increase the clot lysis time in factor VIII (fVIII)-deficient plasma by an activated thrombin-activatable fibrinolysis inhibitor (TAFIa)-dependent mechanism. In the present study, blood was drawn from humans and dogs with hemophilia, and thromboelastography was used to measure tissue factor-initiated fibrin formation and tissue-plasminogen activator-induced fibrinolysis. The kinetics of TAFI and protein C activation by the thrombin-Solulin complex were determined to describe the relative extent of anticoagulation and antifibrinolysis. In severe hemophilia A, clot stability increased by > 4-fold in the presence of Solulin while minimally affecting clot lysis time. Patients receiving fVIII/fIX prophylaxis showed a similar trend of increased clot stability in the presence of Solulin. The catalytic efficiencies of TAFI and protein C activation by the thrombin-Solulin complex were determined to be 1.53 and 0.02/µM/s, respectively, explaining its preference for antifibrinolysis over anticoagulation at low concentrations. Finally, hemophilic dogs given Solulin had improved clot strength in thromboelastography assays. In conclusion, the antifibrinolytic properties of Solulin are exhibited in hemophilic human (in vitro) and dog (in vivo/ex vivo) blood at low concentrations. Our findings suggest the therapeutic utility of Solulin at a range of very low doses.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Enfermedades de los Perros/sangre , Hemofilia A/sangre , Proteínas Recombinantes/farmacología , Adulto , Animales , Enfermedades de los Perros/tratamiento farmacológico , Perros , Fibrinólisis/efectos de los fármacos , Hemofilia A/tratamiento farmacológico , Humanos , Persona de Mediana Edad , Proteolisis/efectos de los fármacos , Receptores de Trombina/uso terapéutico , Proteínas Recombinantes/uso terapéutico , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Tiempo de Coagulación de la Sangre Total , Adulto Joven
14.
Thromb Res ; 128 Suppl 1: S13-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22221845

RESUMEN

Fibrinogen is involved in both primary and secondary hemostasis, playing an important role in platelet aggregation and the establishment of a fibrin network. Recent evidence suggests that very high levels of fibrinogen act as antithrombin and can reduce endogenous thrombin potential and compromise clot stability, particularly following a low tissue factor stimulus. Several laboratory methods for measuring plasma fibrinogen concentrations are available, but results vary depending on the type of method and the use of artificial colloid plasma expanders. Adopting only the Clauss method can provide erroneously high levels when used in patients who have received colloid plasma expanders. This may contribute to a hazardous delay or complete lack of treatment. Multiple in vitro experiments, animal studies, and proof-of-principle randomized, clinical studies have recently suggested that hemostatic intervention with a fibrinogen concentrate may be efficient and safe in controling perioperative bleeding. In particular, fibrinogen concentrate has a key role in improving clotting function and reducing blood loss in settings such as trauma and cardiothoracic surgery. However, prospective studies are needed to determine the clinical efficacy and safety of fibrinogen concentrate when used as a hemostatic intervention for patients with massive bleeding due to trauma or surgery.


Asunto(s)
Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Fibrinógeno/metabolismo , Fibrinógeno/uso terapéutico , Hemostasis , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...