Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 206
Filtrar
1.
Thromb Res ; 237: 37-45, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38547693

RESUMO

BACKGROUND: The hemostatic effect of recombinant (r) factor (F)VIIa after repetitive intermittent administration may be attenuated in patients with hemophilia A (PwHA) with inhibitors (PwHAwI) creating a clinically unresponsive status, although mechanism(s) remain to be clarified. In patients receiving prophylaxis treatment with emicizumab, concomitant rFVIIa is sometimes utilized in multiple doses for surgical procedures or breakthrough bleeding. AIM AND METHODS: We identified 'unresponsiveness' to rFVIIa, based on global coagulation function monitored using rotational thromboelastometry (ROTEM) in 11 PwHAwI and 5 patients with acquired HA, and investigated possible mechanisms focusing on the association between plasma FX levels and rFVIIa-mediated interactions. RESULTS: Our data demonstrated that FX antigen levels were lower in the rFVIIa-unresponsive group than in the rFVIIa-responsive group (0.46 ± 0.14 IU/mL vs. 0.87 ± 0.15 IU/mL, p < 0.01). This relationship was further examined by thrombin generation assays using a FX-deficient PwHAwI plasma model. The addition of FX with rFVIIa was associated with increased peak thrombin (PeakTh) generation. At low levels of FX (<0.5 IU/mL), rFVIIa failed to increase PeakTh to the normal range, consistent with clinical rFVIIa-unresponsiveness. In the presence of emicizumab (50 µg/mL), PeakTh was increased maximally to 80 % of normal, even at low levels of FX (0.28 IU/mL). CONCLUSIONS: Unresponsiveness to rFVIIa was associated with reduced levels of FX in PwHAwI. Emicizumab exhibited in vitro coagulation potential in the presence of FX at concentrations that appeared to limit the clinical response to rFVIIa therapy.

2.
J Thromb Haemost ; 22(2): 441-454, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37926194

RESUMO

BACKGROUND: Factor VIIa induces the release of extracellular vesicles (EVs) from endothelial cells (EEVs). Factor VIIa-released EEVs are enriched with microRNA-10a (miR10a) and elicit miR10a-dependent cytoprotective responses. OBJECTIVES: To investigate mechanisms by which FVIIa induces miR10a expression in endothelial cells and sorts miR10a into the EVs. METHODS: Activation of Elk-1 and TWIST1 expression was analyzed by immunofluorescence microscopy and immunoblot analysis. Small interfering RNA silencing approach was used to knock down the expression of specific genes in endothelial cells. EVs secreted from endothelial cells or released into circulation in mice were isolated by centrifugation and quantified by nanoparticle tracking analysis. Factor VIIa or EVs were injected into mice; mice were challenged with lipopolysaccharides to assess the cytoprotective effects of FVIIa or EVs. RESULTS: FVIIa activation of ERK1/2 triggered the activation of Elk-1, which led to the induction of TWIST1, a key transcription factor involved in miR10a expression. Factor VIIa also induced the expression of La, a small RNA-binding protein. Factor VIIa-driven acid sphingomyelinase (ASM) activation and the subsequent activation of the S1P receptor pathway were responsible for the induction of La. Silencing of ASM or La significantly reduced miR10a levels in FVIIa-released EEVs without affecting the cellular expression of miR10a. Factor VIIa-EEVs from ASM knocked-down cells failed to provide cytoprotective responses in cell and murine model systems. Administration of FVIIa protected wild-type but not ASM-/- mice against lipopolysaccharide-induced inflammation and vascular leakage. CONCLUSION: Our data suggest that enhanced cellular expression of miR10a coupled with La-dependent sorting of miR10a is responsible for enriching FVIIa-released EVs with miR10a.


Assuntos
Vesículas Extracelulares , MicroRNAs , Camundongos , Animais , Fator VIIa/metabolismo , Células Endoteliais/metabolismo , Transdução de Sinais , Lipopolissacarídeos/metabolismo , Vesículas Extracelulares/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo
3.
Biochim Biophys Acta Gen Subj ; 1868(1): 130501, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37925034

RESUMO

BACKGROUND: We previously demonstrated that factor (F)VIII was rapidly activated through proteolytic cleavage at Arg372 and Arg740 by activated FVII (FVIIa)/tissue factor (TF) in very early coagulation phase, followed by inactivation by cleavage at Arg336. The influence of the absence of FVIII B domain in this series of FVIIa/TF-catalyzed reaction remains unclear, however. AIM: To examine the FVIIa/TF-catalyzed reaction of B domain-deleted (BDD-)FVIII. METHODS AND RESULTS: The FVIII activity (FVIII:C) of commercial full-length (FL-)FVIII and BDD-FVIII increased by ∼1.7-fold within 0.5 min after addition of FVIIa/TF (1 nM/0.1 nM). FVIII: C decreased to initial levels with inactivation rate constant (k; ∼0.035) within 15 min of FL-FVIII activation, but decreased gradually to initial levels (k; ∼0.017) within 30 min of BDD-FVIII activation. SDS-PAGE analyses demonstrated that the FVIIa/TF-catalyzed cleavage of BDD-FVIII occurred at Arg336 within 0.5 min in parallel with elevation of FVIII:C, but cleavage at Arg372 was not evident. FVIIa/TF-catalyzed activation of both recombinant BDD-FVIII R336A and R372A mutants that were prepared, were similar to that of wild-type (WT) BDD-FVIII. However, FVIII:C returned to initial levels (k; ∼0.046) within 30 min of R336A mutant activation, but little reduction of FVIII:C was observed with R372A mutant (k; ∼0.0046). SDS-PAGE analysis indicated that FVIIa/TF-catalyzed cleavage of WT and R372A mutant was predominant at Arg336, whereas that of R336A mutant was observed at Arg372. CONCLUSIONS: FVIIa/TF-catalyzed activation of BDD-FVIII was initiated by cleavage at Arg336, and the FVIII B domain appeared to control FVIIa/TF-catalyzed reactions by altering pattern of cleavage at Arg336 and Arg372.


Assuntos
Fator VIIa , Tromboplastina , Fator VIII/genética , Coagulação Sanguínea , Catálise
4.
J Thromb Haemost ; 21(12): 3414-3431, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37875382

RESUMO

BACKGROUND: Our recent studies showed that activated factor (F) VII (FVIIa) releases extracellular vesicles (EVs) from the endothelium. FVIIa-released EVs were found to be enriched with phosphatidylserine (PS) and contribute to the hemostatic effect of FVIIa in thrombocytopenia and hemophilia. OBJECTIVE: To investigate mechanisms by which FVIIa induces EV biogenesis and enriches EVs with PS. METHODS: FVIIa activation of acid sphingomyelinase (aSMase) was evaluated by its translocation to the cell surface. The role of aSMase in the biogenesis of FVIIa-induced EVs and their enrichment with PS was investigated using specific siRNAs and inhibitors of aSMase and its downstream metabolites. Wild-type and aSMase-/- mice were injected with a control vehicle or FVIIa. EVs released into circulation were quantified by nanoparticle tracking analysis. EVs hemostatic potential was assessed in a murine thrombocytopenia model. RESULTS: FVIIa activation of aSMase is responsible for both the externalization of PS and the release of EVs in endothelial cells. FVIIa-induced aSMase activation led to ceramide generation and de novo expression of transmembrane protein 16F. Inhibitors of ceramidases, sphingosine kinase, or sphingosine-1-phosphate receptor modulator blocked FVIIa-induced expression of transmembrane protein 16F and PS externalization without interfering with FVIIa release of EVs. In vivo, FVIIa release of EVs was markedly impaired in aSMase-/- mice compared with wild-type mice. Administration of a low dose of FVIIa, sufficient to induce EVs release, corrected bleeding associated with thrombocytopenia in wild-type mice but not in aSMase-/- mice. CONCLUSION: Our study identifies a novel mechanism by which FVIIa induces PS externalization and releases PS-enriched EVs.


Assuntos
Vesículas Extracelulares , Hemostáticos , Trombocitopenia , Animais , Camundongos , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Fator VIIa/metabolismo , Fosfatidilserinas/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Trombocitopenia/metabolismo
5.
Front Oncol ; 13: 1232621, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37546403

RESUMO

Diffuse alveolar hemorrhage (DAH) is a life-threatening complication of hematopoietic cellular therapy (HCT). This study aimed to evaluate the effect of DAH treatments on outcomes using data from consecutive HCT patients clinically diagnosed with DAH from 3 institutions between January 2018-August 2022. Endpoints included sustained complete response (sCR) defined as bleeding cessation without recurrent bleeding, and non-relapse mortality (NRM). Forty children developed DAH at a median of 56.5 days post-HCT (range 1-760). Thirty-five (88%) had at least one concurrent endothelial disorder, including transplant-associated thrombotic microangiopathy (n=30), sinusoidal obstructive syndrome (n=19), or acute graft versus host disease (n=10). Fifty percent had a concurrent pulmonary infection at the time of DAH. Common treatments included steroids (n=17, 25% sCR), inhaled tranexamic acid (INH TXA,n=26, 48% sCR), and inhaled recombinant activated factor VII (INH fVIIa, n=10, 73% sCR). NRM was 56% 100 days after first pulmonary bleed and 70% at 1 year. Steroid treatment was associated with increased risk of NRM (HR 2.25 95% CI 1.07-4.71, p=0.03), while treatment with INH TXA (HR 0.43, 95% CI 0.19- 0.96, p=0.04) and INH fVIIa (HR 0.22, 95% CI 0.07-0.62, p=0.005) were associated with decreased risk of NRM. Prospective studies are warranted to validate these findings.

6.
J Extra Corpor Technol ; 55(2): 60-69, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37378438

RESUMO

BACKGROUND: Neonates and infants undergoing cardiac surgery tend to receive high volumes of blood products. The use of rotational thromboelastometry (ROTEM®) has been shown to reduce the administration of blood products in adults after cardiac surgery. We sought to develop a targeted administration of blood products based on ROTEM® to reduce blood product utilization during and after neonatal and infant cardiac surgery. METHODS: We conducted a retrospective review of data from a single center for neonates and infants undergoing congenital cardiac surgery using cardiopulmonary bypass (CPB) from September 2018-April 2019 (control group). Then, using a ROTEM® algorithm, we collected data prospectively between April-November 2021 (ROTEM group). Data collected included age, weight, gender, procedure, STAT score, CPB time, aortic cross-clamp time, volume, and type of blood products administered in the operating room and cardiothoracic intensive care unit (CTICU). In addition, ROTEM® data, coagulation profile in CTICU, chest tube output at 6 and 24 hours, use of factors concentrate, and thromboembolic complications were recorded. RESULTS: The final cohort of patients included 28 patients in the control group and 40 patients in the ROTEM group. The cohort included neonates and infants undergoing the following procedures: arterial switch, aortic arch augmentation, Norwood procedure, and comprehensive stage II procedure. There were no differences in the demographics or procedure complexity between the two groups. Patients in the ROTEM® group received fewer platelets (36 ± 12 vs. 49 ± 27 mL/kg, p 0.028) and cryoprecipitate (8 ± 3 vs. 15 ± 10 mL/kg, p 0.001) intraoperatively when compared to the control group. CONCLUSION: The utilization of ROTEM® may have contributed to a significant reduction in some blood product administration during cardiac surgery for infants and neonates. ROTEM® data may play a role in reducing blood product administration in neonatal and infant cardiac surgery.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Recém-Nascido , Adulto , Humanos , Lactente , Procedimentos Cirúrgicos Cardíacos/métodos , Testes de Coagulação Sanguínea , Tromboelastografia/métodos , Estudos Retrospectivos , Algoritmos
7.
Ann Hematol ; 2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-37391649

RESUMO

rFVIIa, a human recombinant activated coagulation factor VII, has been used worldwide for more than two decades for the treatment of bleeding episodes and prevention of bleeding in patients undergoing surgery/invasive procedures with congenital haemophilia A or B with inhibitors (CHwI A or B), acquired haemophilia (AH), congenital factor VII deficiency and Glanzmann thrombasthenia (GT), refractory to platelet transfusion. The approved dosage, administration and indication of rFVIIa in the US, Europe and Japan differ, depending on the needs of the patient population and regulatory practices. This review presents an overview of the current status and future prospects, including that from a Japanese perspective, of using rFVIIa in the treatment of approved indications. The efficacy and safety of rFVIIa in the approved indications has been demonstrated in several randomised and observational studies and data from registries. The overall incidence of thrombosis across all approved indications in a retrospective safety assessment of clinical trials and registries, prelicensure studies and postmarketing surveillance studies of rFVIIa use was 0.17%. Specifically, the risk of thrombotic events was 0.11% for CHwI, 1.77% for AH, 0.82% for congenital factor VII deficiency and 0.19% for GT. Emerging non-factor therapies such as emicizumab have changed the treatment landscape of haemophilia A, including preventing bleeding in patients with CHwI. However, rFVIIa will continue to play a significant role in the treatment of such patients, particularly during breakthrough bleeding or surgical procedures.

8.
J Thromb Haemost ; 21(6): 1493-1502, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37037699

RESUMO

BACKGROUND: Mim8 is a novel antifactor IXa/antifactor X bispecific antibody in clinical development for prophylactic treatment of hemophilia A with and without inhibitors. Patients treated with Mim8 may need supplementary bleed treatment under certain conditions such as surgery or major trauma. OBJECTIVES: This study aimed to better understand the response of Mim8 in thrombin generation assays (TGAs) alone or in combination with other hemostatic proteins. METHODS: We used TGAs with different activators (tissue factor (TF) and activated factor XI) to better understand the similarities and differences between the mode of action of Mim8 and factor VIII (FVIII). Following this, we investigated the effects of mixing Mim8 with the main bleed treatment options for persons with hemophilia A with or without inhibitors: FVIII, activated factor VII (FVIIa), and activated prothrombin complex concentrates (aPCC). RESULTS: The results indicated that for patients without inhibitors, Mim8 does not interfere with FVIII's mode of action. For patients with inhibitors, Mim8 mixed with aPCC results in a strong synergistic effect causing thrombin generation far exceeding the normal levels. Contrary to this, mixing Mim8 with FVIIa results in a more controlled additive effect, visible only when using TF as a trigger, which does not exceed the normal level of thrombin generation. CONCLUSION: These findings support the use of approved clinical doses of FVIIa for bleed treatment of patients with FVIII inhibitors treated with Mim8. Additionally, the findings suggest that concomitant use of FVIII and Mim8 is safe for managing breakthrough bleeds.


Assuntos
Hemofilia A , Hemostáticos , Humanos , Fator VIII , Trombina/metabolismo , Fator VIIa/metabolismo , Hemostáticos/uso terapêutico , Hemorragia/tratamento farmacológico , Fator IX/uso terapêutico , Tromboplastina/uso terapêutico , Proteínas Recombinantes/uso terapêutico
9.
J Med Case Rep ; 17(1): 161, 2023 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-37055848

RESUMO

BACKGROUND: The clotting or hemostasis system is a meticulously regulated set of enzymatic reactions that occur in the blood and culminate in formation of a fibrin clot. The precisely calibrated signaling system that prevents or initiates clotting originates with the activated Factor Seven (FVIIa) complexed with tissue factor (TF) formed in the endothelium. Here we describe a rare inherited mutation in the FVII gene which is associated with pathological clotting. CASE PRESENTATION: The 52-year-old patient, with European, Cherokee and African American origins, FS was identified as having low FVII (10%) prior to elective surgery for an umbilical hernia. He was given low doses of NovoSeven (therapeutic Factor VIIa) and had no unusual bleeding or clotting during the surgery. In fact, during his entire clinical course he had no unprovoked bleeding. Bleeding instances occurred with hemostatic stresses such as gastritis, kidney calculus, orthopedic surgery, or tooth extraction, and these were handled without factor replacement. On the other hand, FS sustained two unprovoked and life-threatening instances of pulmonary emboli, although he was not treated with NovoSeven at any time close to the events. Since 2020, he has been placed on a DOAC (Direct Oral Anticoagulant, producing Factor Xa inhibition) and has sustained no further clots. POSSIBLE MECHANISM OF (UNAUTHORIZED) FVII ACTIVATION: FS has a congenitally mutated FVII/FVIIa gene, which carries a R315W missense mutation in one allele and a mutated start codon (ATG to ACG) in the other allele, thus rendering the patient effectively homozygous for the missense FVII. Structure based comparisons with known crystal structures of TF-VIIa indicate that the patient's missense mutation is predicted to induce a conformational shift of the C170's loop due to crowding of the bulky tryptophan to a distorted "out" position (Fig. 1). This mobile loop likely forms new interactions with activation loop 3, stabilizing a more active conformation of the FVII and FVIIa protein. The mutant form of FVIIa may be better able to interact with TF, displaying a modified serine protease active site with enhanced activity for downstream substrates such as Factor X. CONCLUSIONS: Factor VII can be considered the gatekeeper of the coagulation system. Here we describe an inherited mutation in which the gatekeeper function is altered. Instead of the expected bleeding manifestations resulting from a clotting factor deficiency, the patient FS suffered clotting episodes. The efficacy of the DOAC in treating and preventing clots in this unusual situation is due to its target site of inhibition (anti-Xa), which lies downstream of the site of action of FVIIa/TF.


Assuntos
Fator VIIa , Trombose , Humanos , Pessoa de Meia-Idade , Fator VIIa/uso terapêutico , Fator VIIa/química , Fator VIIa/metabolismo , Alelos , Tromboplastina/química , Tromboplastina/metabolismo , Coagulação Sanguínea/genética , Trombose/tratamento farmacológico , Modelos Estruturais
10.
J Clin Med ; 12(8)2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37109356

RESUMO

BACKGROUND: Hemostasis in critically ill patients represents a fragile balance between hypocoagulation and hypercoagulation, and is influenced by various factors. Perioperative use of extracorporeal membrane oxygenation (ECMO)-increasingly used in lung transplantation-further destabilizes this balance, not least due to systemic anticoagulation. In the case of massive hemorrhage, guidelines recommend considering recombinant activated Factor VII (rFVIIa) as an ultima ratio treatment only after several preconditions of hemostasis have been established. These conditions are calcium levels ≥ 0.9 mmol/L, fibrinogen levels ≥ 1.5 g/L, hematocrit ≥ 24%, platelet count ≥ 50 G/L, core body temperature ≥ 35 °C, and pH ≥ 7.2. OBJECTIVES: This is the first study to examine the effect of rFVIIa on bleeding lung transplant patients undergoing ECMO therapy. The fulfillment of guideline-recommended preconditions prior to the administration of rFVIIa and its efficacy alongside the incidence of thromboembolic events were investigated. METHODS: In a high-volume lung transplant center, all lung transplant recipients receiving rFVIIa during ECMO therapy between 2013 and 2020 were screened for the effect of rFVIIa on hemorrhage, fulfillment of recommended preconditions, and incidence of thromboembolic events. RESULTS AND DISCUSSION: Of the 17 patients who received 50 doses of rFVIIa, bleeding ceased in four patients without surgical intervention. Only 14% of rFVIIa administrations resulted in hemorrhage control, whereas 71% of patients required revision surgery for bleeding control. Overall, 84% of all recommended preconditions were fulfilled; however, fulfillment was not associated with rFVIIa efficacy. The incidence of thromboembolic events within five days of rFVIIa administration was comparable to cohorts not receiving rFVIIa.

11.
World J Pediatr Congenit Heart Surg ; 14(3): 282-288, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36919404

RESUMO

OBJECTIVE: This study aims to compare the efficacy and safety of activated recombinant factor VII (rFVIIa) and prothrombin complex concentrate (PCC) in the treatment of bleeding complications following surgery requiring cardiopulmonary bypass (CPB) in children. DESIGN/METHODS: This is a retrospective chart review of a single institution comprising patients aged 0 to 18 years old with congenital heart disease. Patients must have received either PCC or rFVIIa after coming off CPB. Our primary efficacy endpoint is time in the operating room from off-CPB to pediatric intensive care unit admission. Our primary safety endpoint is thrombosis through 30 days. RESULTS: Our primary efficacy outcome was significantly shorter in the PCC group compared with the rFVIIa group (P < .0001). Similarly, secondary efficacy outcomes of packed red blood cell administration, chest tube output, and transfusion exposures all significantly favored PCC administration. However, CPB time was significantly longer, and body temperatures were significantly lower, in the rFVIIa group. Safety outcomes, including our primary safety outcome of thrombosis through 30 days, were similar between the two groups. CONCLUSION: This study questions whether PCC could be favored over rFVIIa for hemostasis in children with congenital heart disease following CPB surgery. In addition, this study has found no difference when comparing PCC and rFVIIa in terms of safety outcomes, particularly thrombosis events. There are several limitations to this study due to the retrospective nature of the design and the differences between the two study groups. Despite the limitations, this study suggests that relatively early administration of PCC could be favored over delayed administration of rFVIIa to control recalcitrant post-CPB bleeding in the operating room.


Assuntos
Fator VIIa , Trombose , Humanos , Criança , Recém-Nascido , Lactente , Pré-Escolar , Adolescente , Fator VIIa/uso terapêutico , Ponte Cardiopulmonar/efeitos adversos , Estudos Retrospectivos , Hemorragia/tratamento farmacológico , Hemorragia/etiologia , Trombose/tratamento farmacológico , Trombose/etiologia , Proteínas Recombinantes/uso terapêutico
13.
J Thromb Haemost ; 21(4): 917-932, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36696201

RESUMO

BACKGROUND: Tissue factor (TF), a transmembrane glycoprotein, plays a profound role in the formation of the tissue factor-factor VIIa (TF-FVIIa) complex that initiates factor Xa (FXa) generation followed by thrombin activation and clot formation. Previous reports suggest that TF-FVIIa coagulant activity at the cell surface may be affected by various processes, including changes in cholesterol content and posttranslational modifications of TF. Numerous studies were conducted but yielded inconclusive results about the effect of cholesterol on TF expression. OBJECTIVE: The present study aimed to understand how cholesterol affects structural modulations on the tissue factor-factor VIIa-factor Xa ternary complex (TF-FVIIa-FXa). Additionally, we aimed to illustrate the effect of palmitoylation on the Cys245 residue of TF and understand its structural implications on the TF-FVIIa-FXa. METHODS: We set up the following 4 systems in different lipid environments: TF-FVIIa-FXa in POPC:POPS (CS), TF-FVIIa-FXa in POPC:POPS:CHOL (CSL), Palmitoylated TF-FVIIa-FXa in POPC:POPS:CHOL (CSLP), and Palmitoylated TF-FVIIa-FXa in POPC:CHOL (CLP), respectively, and subjected them to molecular dynamics simulation. RESULTS: Hydrogen-bond and contact probability analysis were performed between various important domains of TF-FVIIa-FXa and notable novel interactions: Asn93FVIIa:L-Lys48TF, Arg178FVIIa:H-Asp95FXa:B, Lys20FVIIa:H-Glu193FXa:A, Arg178FVIIa:H-Asp97FXa:B, and Arg153FVIIa:H-Gln135FXa:B have been reported. The protein stability study implies that the CS and CLP systems are thermodynamically less stable than CSL and CSLP systems. CONCLUSION: Analysis of molecular dynamic simulation data suggests that the presence of cholesterol and palmitoylation may contribute to structural rigidity, stability, and compactness of key domains of TF-FVIIa-FXa by augmenting protein-protein and protein-lipid interactions.


Assuntos
Fator Xa , Tromboplastina , Humanos , Fator VIIa/química , Fator VIIa/metabolismo , Fator Xa/química , Fator Xa/metabolismo , Lipídeos/química , Lipoilação , Simulação de Dinâmica Molecular , Tromboplastina/química , Tromboplastina/metabolismo , Colesterol/química , Colesterol/metabolismo
14.
Vnitr Lek ; 68(8): 498-507, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36575067

RESUMO

Haemophagocytic syndrome, diffuse alveolar haemorrhage, catastrophic antiphospholipid syndrome and various types of thrombotic microangiopathies are rare conditions with significant morbidity and mortality. A common feature is late diagnosis, which can affect the success of treatment. The aim of this review article is to summarize the basic diagnostic and therapeutic steps of the present subpopulation of critically ill patients.


Assuntos
Síndrome Antifosfolipídica , Linfo-Histiocitose Hemofagocítica , Microangiopatias Trombóticas , Humanos , Adulto , Microangiopatias Trombóticas/diagnóstico , Síndrome Antifosfolipídica/complicações , Síndrome Antifosfolipídica/diagnóstico , Síndrome Antifosfolipídica/terapia
15.
Thromb Res ; 215: 58-66, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35640516

RESUMO

INTRODUCTION: Acute bleeding leads to significant morbidity and mortality. Recombinant wildtype Factor VIIa (WT FVIIa) had been reported to have some therapeutic effects in some clinical trials, however, its use was associated with thromboembolic events. We sought to develop a novel FVIIa molecule (CT-001) with enhanced activity and lowered thrombogenicity risk. METHODS AND METHODS: CT-001 has 4 N-glycans (T106N/N145/V253N/N322) with terminal sialic acid residues removed to promote active clearance via the asialoglycoprotein receptor, and P10Q/K32E substitutions introduced to its gamma-carboxyglutamic acid (Gla) domain for enhanced phospholipid affinity and activity. RESULTS: In mice, CT-001 had a half-life of 5 min and a clearance of 467 mL/h/kg at 3 mg/kg, significantly faster than WT FVIIa (t1/2 = 1.8 h, Cl = 39 mL/h/kg). Interestingly, CT-001 was efficacious in reducing blood loss even with its rapid clearance. In a severe hemorrhage mouse model with tail amputated 5 cm from the tip, 1 mg/kg CT-001 provided efficacy comparable to 3 mg/kg WT FVIIa. The fast clearance of CT-001 resulted in significantly reduced thrombogenicity in comparison to WT FVIIa in a FeCl3-induced carotid artery thrombosis mouse model, and further confirmed in a soluble tissue factor-induced thrombosis model. CONCLUSIONS: The data on CT-001 demonstrate that a short duration of highly active FVIIa procoagulant activity has the potential to be an optimal paradigm for the treatment of acute bleeds.


Assuntos
Fator VIIa , Hemostáticos , Animais , Modelos Animais de Doenças , Fator VIIa/farmacologia , Fator VIIa/uso terapêutico , Hemorragia/tratamento farmacológico , Humanos , Camundongos , Tromboplastina , Tomografia Computadorizada por Raios X
17.
Medicines (Basel) ; 9(3)2022 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-35323720

RESUMO

Acquired hemophilia A (AHA) is a rare disease with a prevalence in Europe of 1.5 per million. This diagnosis is significantly delayed in about one-third of all cases, leading to deferred treatment. The main signs of AHA are spontaneous bleeding seen in about two-thirds of all patients. AHA can be lethal in 20% of all symptomatic cases. This patient population's main standard laboratory finding is a prolonged aPTT (activated prothrombin Time) with otherwise normal coagulation results. In addition, antibodies against FVIII (in Bethesda Units) and a quantitative reduction of FVIII activity are necessary to confirm AHA. The therapy of acute bleeding related to AHA is based on the following main principles: Pharmacologic control of the bleeding is of absolute importance. It can be achieved by administering either recombinant activated FVIIa "bypass therapy"; activated prothrombin complex; or Emicizumab, a bispecific monoclonal antibody. Eradication of the FVIII antibodies should be initiated simultaneously. The combination of steroids with cyclophosphamide leads to the highest eradication rates. Causes of AHA may be related to neoplasms, autoimmune diseases, and pregnancy. We report on a patient who underwent four surgical procedures before the diagnosis of AHA was established.

18.
Drugs Today (Barc) ; 58(3): 105-116, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35274630

RESUMO

Hemophilia A and B are X-linked hereditary bleeding disorders due to factor VIII (FVIII) or factor IX (FIX) deficiency, respectively. Major advancements have been made in the care of patients with hemophilia, yet the development of inhibitors to infused FVIII or FIX continues to be a formidable challenge. The current first-line therapy for acute bleeding episodes in patients diagnosed with inhibitors are bypassing agents including activated prothrombin complex concentrates (aPCCs) and recombinant factor VIIa (rFVIIa). Eptacog beta (SevenFact; LFB Biotechnologies, Hema Biologics) is a new rFVIIa product produced via expression in the milk of transgenic rabbits. This emerging platform has demonstrated numerous cost advantages to traditional cell culture systems including a better ability to scale up production and better protein yields. Eptacog beta is currently approved by the U.S. Food and Drug Administration (FDA) for the on-demand control of bleeding episodes in patients with hemophilia aged 12 to 75 with inhibitors. A potential future expansion of its current label could occur given the recent completion of two major phase III clinical trials evaluating its efficacy in children as well as its use for perioperative management. In this paper, we describe the preclinical and clinical literature documenting the development of eptacog beta and discuss its current and future application for the management of patients with hemophilia and inhibitors.


Assuntos
Fator VIIa , Hemofilia A , Hemofilia B , Proteínas Recombinantes , Animais , Fator VIIa/uso terapêutico , Hemofilia A/tratamento farmacológico , Hemofilia B/tratamento farmacológico , Humanos , Coelhos , Proteínas Recombinantes/uso terapêutico
19.
Scand J Clin Lab Invest ; 82(2): 156-161, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35175155

RESUMO

The Danish Capital Region Blood Bank operates a 24/7 on-call service staffed with physicians specialized in hemostatic management to guide clinicians in hemostatic resuscitation, including administration of prohemostatic therapy (PHT). The outcome of patients who receive PHT as part of hemostatic resuscitation remains unanswered. The objective of this study was therefore to investigate clinical outcome of patients receiving PHT managed by the on-call service. We identified 287 patients who received PHT during 2015-16, of which 161 (59%) received fibrinogen concentrate (FC), 111 (39%) received prothrombin complex concentrate (PCC), and 15 (5%) received recombinant factor VIIa (rFVIIa) as the first product. Patients were critically ill with a 30-day mortality of 31%. Among FC recipients, cardiothoracic admission, non-trauma, and antithrombotics predicted survival. FC recipients had lower platelet count and thrombelastography clot strengths than the other PHT groups and within the group, these factors predicted mortality. The symptomatic thromboembolic event (TE) rate at 30 days was 5%. For PCC recipients, vitamin K antagonists predicted survival, while rivaroxaban predicted mortality. TE rate was 2%. We did not identify factors associated with survival in the small group of rFVIIa recipients. TE rate was 13%. In summary, trauma and coagulopathy predicted mortality in patients who received FC and our data suggest that optimization of PHT algorithms may be possible. Outcome of patients who received PCC was comparable to results reported elsewhere and its use may be safe in a setting as reported here. Recombinant FVIIa was rarely used but had the highest incidence of arterial thromboembolism.


Assuntos
Hemostáticos , Tromboembolia , Fatores de Coagulação Sanguínea/uso terapêutico , Fator VIIa , Fibrinogênio/análise , Fibrinogênio/uso terapêutico , Objetivos , Hemostáticos/uso terapêutico , Humanos , Proteínas Recombinantes/efeitos adversos , Estudos Retrospectivos , Tromboembolia/tratamento farmacológico
20.
J Reprod Immunol ; 150: 103473, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35030354

RESUMO

Placenta is a crucial source of Tissue Factor (TF) to initiate coagulation. As far as the TF is concern, aberrant expression of TF has been reported to have a significant role in thrombosis, inflammation, cancer metastasis and atherosclerosis. It is evident that TF and TF-FVIIa complex has major roles in the disease process beyond hemostasis and thrombosis. On the other hand, TF-FVII-dependent signaling primarily activates PAR2 and inducing pro-angiogenic and immune-modulating cytokines in tumor environment. However, the role of TF has not been delineated in placental functions. Integrin typically binds to the extracellular matrix which in turn mediate cell-cell adhesion and cell behavior for migration. Dysregulation of integrin expression affects cell interaction, proliferation, and migration. Therefore, this study aims to ascertain the expression of TF in HTR-8/SVneo trophoblast cell line and its role in signal transduction of integrin (ITGα1, ITGα2, ITGß1) regulation concerning the invasion of trophoblasts. We have used RT-PCR and Western blot for the gene and protein expression analysis respectively. In addition, cell migration assays, MTT, and DAPI were performed to examine migration, cytotoxicity and apoptosis effect of FVIIa. The results suggest that the gene and protein level expressions of TF were predominant in HTR-8/SVneo cell line. Further, the cytotoxicity and apoptosis in HTR-8/SVneo cells were not observed when treated with FVIIa. The cells treated with FVIIa shown a dose-dependent up-regulation of integrin(s) (**p < 0.01, *p < 0.05) when compared to control. Migration of the HTR-8/SVneo cells was observed without any apoptosis in FVIIa-treated cells when compared to that of control. On the whole, these observations delineated the TF-FVIIa interaction in modulating the TF-dependent integrin signal transduction in HTR-8/SVneo trophoblast cell line.


Assuntos
Tromboplastina , Trofoblastos , Movimento Celular , Feminino , Humanos , Integrinas/metabolismo , Placenta/metabolismo , Gravidez , Tromboplastina/genética , Tromboplastina/metabolismo , Trofoblastos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...