Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
J Thromb Haemost ; 22(7): 1857-1866, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38583717

ABSTRACT

BACKGROUND: Emicizumab has been approved for the prophylaxis of patients with hemophilia A with or without inhibitors. However, spontaneous and trauma-induced breakthrough bleeds have been reported in patients on emicizumab prophylaxis, and no laboratory assay has been validated to evaluate the hemostatic activity of emicizumab. OBJECTIVES: The thrombin generation assay (TGA) could be a surrogate marker of the hemostatic efficacy of emicizumab. The correlation between TGA and the methods used to measure emicizumab blood concentration was evaluated in this study. METHODS: TGA was modified by the use of a trigger reagent combining a very low concentration of tissue factor and activated factor (F)XI. Emicizumab quantification was performed by 3 methods: the modified 1-step FVIII assay and 2 methods based on liquid chromatography and mass spectrometry (LC-MS). RESULTS: Using tissue factor/activated FXI-triggered TGA and platelet-poor plasma, a relationship was observed between the area under the thrombin generation curve (endogenous thrombin potential [ETP]) and the clinical response of patients to emicizumab. The ultrastructure of fibrin clots was consistent with ETP results and showed that emicizumab had a hemostatic activity equivalent to 20 to 30 IU/dL of FVIII. Finally, pharmacokinetic/pharmacodynamic analyses showed no correlation between ETP and LC-MS nor with modified 1-stage FVIII assay, but a statistically significant correlation between the LC-MS methods and the time-to-peak results of the TGA. CONCLUSION: Using a modified TGA, this study showed that patients who experienced breakthrough bleeds while on emicizumab had a lower thrombin-generating capacity compared with others with good clinical response to emicizumab.


Subject(s)
Antibodies, Bispecific , Antibodies, Monoclonal, Humanized , Hemophilia A , Hemostasis , Thrombin , Humans , Hemophilia A/drug therapy , Hemophilia A/blood , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/pharmacokinetics , Antibodies, Bispecific/therapeutic use , Antibodies, Bispecific/blood , Thrombin/metabolism , Adult , Hemostasis/drug effects , Male , Young Adult , Adolescent , Hemorrhage/blood , Middle Aged , Treatment Outcome , Chromatography, Liquid , Blood Coagulation Tests , Hemostatics/therapeutic use , Hemostatics/pharmacology , Hemostatics/pharmacokinetics , Mass Spectrometry , Blood Coagulation/drug effects , Child , Severity of Illness Index
2.
J Thromb Haemost ; 22(4): 990-1000, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38142846

ABSTRACT

BACKGROUND: Mim8 (denecimig) is a factor VIII (FVIII) mimetic bispecific antibody in development for the treatment of hemophilia. Data from the phase 1 part of FRONTIER1 (EudraCT: 2019-000465-20, NCT04204408, and NN7769-4513) suggested that Mim8 was well tolerated in healthy participants and exhibited pharmacokinetic (PK) properties consistent with dose proportionality. OBJECTIVES: The partially randomized, phase 2, multiple ascending dose (MAD) part of FRONTIER1 aimed to evaluate the safety, PK, pharmacodynamics (PD), and exploratory efficacy of Mim8 in participants with hemophilia A with or without FVIII inhibitors. METHODS: The MAD part of FRONTIER1 consisted of 42 participants, assigned to 5 cohorts, with participants in cohorts 3 and 4 randomized 1:1 to dosing weekly or every 4 weeks, respectively. Four of the 42 participants (9.5%) had FVIII inhibitors prior to study enrolment. The primary endpoint was treatment-emergent adverse events (TEAEs). PK and PD were evaluated by Mim8 plasma concentration and thrombin generation, respectively. Exploratory efficacy was assessed via the number of treated bleeds. Safety and PD parameters were also evaluated from an exploratory cohort treated with emicizumab. RESULTS: Mim8 was well tolerated, with 1 serious TEAE (anxiety-related chest pain) deemed unrelated to Mim8. There was no dose dependency on the number, causality, type, or severity of TEAEs. PK/PD properties supported weekly to monthly dosing approaches, and few participants experienced treated bleeds beyond the lowest dose cohort (1 in cohorts 2 and 3, and 3 in cohort 5). CONCLUSION: These data support the continued clinical development of Mim8, and FRONTIER1 has proceeded onto an extension phase.


Subject(s)
Hemophilia A , Hemostatics , Humans , Factor VIIIa/adverse effects , Factor VIIIa/pharmacokinetics , Factor VIIIa/pharmacology , Hemophilia A/diagnosis , Hemophilia A/drug therapy , Hemorrhage/drug therapy , Hemostatics/adverse effects , Hemostatics/pharmacokinetics , Hemostatics/pharmacology , Thrombin
3.
Thromb Haemost ; 122(5): 715-725, 2022 05.
Article in English | MEDLINE | ID: mdl-34407556

ABSTRACT

BACKGROUND: The asialoglycoprotein receptor (ASGPR) binds with high affinity factor VIII (FVIII) through its N-linked oligosaccharides. However, its contribution to the wide inter-individual variation of infused FVIII pharmacokinetics (PK) in hemophilia A (HA) is unknown. OBJECTIVE: To investigate the variability in FVIII PK outcomes in relation to genetic variation in the ASGR2, encoding the ASGPR2 subunit. METHODS: Thirty-two HA patients with FVIII:C ≤2 IU/dL underwent 66 single-dose FVIII PK studies. PK parameters were evaluated in relation to ASGR2 5' untranslated region (5'UTR) polymorphisms, which were investigated by recombinant and white blood cell reverse transcription-polymerase chain reaction approaches. RESULTS: The 5'UTR polymorphisms determine a frequent and conserved haplotype (HT1) in a regulatory region. The HT1 homozygotes may differ in the amounts of alternatively spliced mRNA transcripts and thus ASGPR2 isoforms. Compared with the other ASGR2 genotypes, the c.-95TT homozygotes (n = 9), showed threefold longer Alpha HL (3.60 hours, 95% confidence interval: 1.44-5.76, p = 0.006), and the c.-95TC heterozygotes (n = 17) showed 25% shorter mean residence time (MRT; 18.5 hours, 15.0-22.0, p = 0.038) and 32% shorter Beta HL (13.5 hours, 10.9-16.0, p = 0.016). These differences were confirmed in patients (n = 27) undergoing PK studies (n = 54) with full-length FVIII only. In different linear regression models, the contribution of the ASGR2 genotypes remained significant after adjustment by ABO genotypes and von Willebrand factor (VWF) antigen levels, and explained 14% (MRT), 15 to 18% (Beta HL), and 22% (Alpha HL) of parameter variability. CONCLUSION: Infused FVIII distribution was modulated by frequent ASGR2 genotypes, independently from and together with ABO and VWF antigen levels, which has potential implications for genetically tailored substitutive treatment in HA.


Subject(s)
Asialoglycoprotein Receptor , Factor VIII , Hemophilia A , Hemostatics , 5' Untranslated Regions , Asialoglycoprotein Receptor/genetics , Factor VIII/pharmacokinetics , Hemophilia A/drug therapy , Hemophilia A/genetics , Hemostatics/pharmacokinetics , Humans , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
4.
Haemophilia ; 27(4): 626-633, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33966319

ABSTRACT

INTRODUCTION: The use of pharmacokinetic assessment for optimal prophylactic dosing of factor concentrates in haemophilia has gained increasing enthusiasm over the last decade. However, blood sampling on several occasions is burdensome and limited sampling using population-based PK is appealing. AIM: To compare the pharmacokinetics and dosing recommendations for prophylaxis using six-point single subject versus population-based method (WAPPS-Hemo) for simoctocog alfa (Nuwiq® ). METHODS: Twelve adult patients with severe haemophilia A received a factor VIII (FVIII) dose of ≈50 IU/kg, and the activity was measured pre-infusion and at 30 min, 6, 9, 24 and 48 h post-infusion. Half-life (t1 /2 ), weight-normalized AUC and time to troughs of 5%, 3% and 1% were calculated. The correlation between the PK algorithms was assessed using intraclass correlations (ICC) and dosing estimations were provided. RESULTS: WAPPS-Hemo yielded a slightly longer mean t1 /2 , but the overall correlation between the methods was good (ICC ≥0.79) The time to troughs of 5%, 3% and 1% showed ICCs ≥0.86. For all variables, the most converging limited time point was 6+48 h. Additional time points did not improve the correlation. Despite similar pharmacokinetics, the mean estimated dose for a specific trough level varied from 60% less to 20% more using the population-based approach. The time to 1% and the corresponding dose was sensitive to the baseline assumption. CONCLUSION: Our data support the use of population-based PK for patients on simoctocog alfa prophylaxis but also indicates differences, stressing the importance of the sampling scheme and monitoring actual FVIII levels achieved.


Subject(s)
Factor VIII , Hemophilia A , Hemostatics , Adult , Blood Specimen Collection , Factor VIII/pharmacokinetics , Factor VIII/therapeutic use , Half-Life , Hemophilia A/drug therapy , Hemostatics/pharmacokinetics , Hemostatics/therapeutic use , Humans
5.
Carbohydr Polym ; 264: 117965, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33910708

ABSTRACT

Hemostasis is of great significance regardless of the smooth operation or postoperative recovery. Therefore, it is urgent to develop a hemostatic material with excellent biodegradability and biocompatibility. It is well known that both carboxymethyl chitosan and hyaluronic acid with biodegradability and biocompatibility have wound healing promoting property. Here, a degradable chitosan-based hydrogel was prepared based on carboxymethyl chitosan and cross-linked by oxidized hyaluronic acid. The hemostatic performance of the hydrogel in rat liver resection injury was evaluated which results showed that the hydrogel exhibited comparable hemostatic properties compared with Fibrin Sealant. In addition, the hydrogel proved to be rapidly absorbed by the body without significant accumulation in vivo, demonstrating good biodegradability and biocompatibility. The overall results suggested the hydrogel will be a promising hemostatic hydrogel for controlling bleeding.


Subject(s)
Chitosan/pharmacokinetics , Hemostatics/pharmacokinetics , Hyaluronic Acid/pharmacokinetics , Hydrogels/pharmacokinetics , Wounds and Injuries/therapy , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacokinetics , Chitosan/analogs & derivatives , Chitosan/chemistry , Chitosan/metabolism , Cross-Linking Reagents/chemistry , Female , Hemostasis , Hemostatics/chemistry , Hemostatics/metabolism , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Hydrogels/chemistry , Hydrogels/metabolism , Liver/metabolism , Male , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Tissue Distribution , Wound Healing/drug effects , Wounds and Injuries/metabolism
6.
J Biomater Appl ; 35(8): 978-993, 2021 03.
Article in English | MEDLINE | ID: mdl-33269628

ABSTRACT

Polymers derived from natural sources are of interest in the scientific and medical communities, especially soy protein which exhibits low immunogenicity and good mechanical properties, and supports cell proliferation. Soy protein is cost-effective compared to other natural polymers and is attractive also due to its non-animal origin and relatively long storage stability. In the current study, hybrid film structures were developed and studied as a novel wound dressing platform with controlled release of three bioactive agents. The dense top layer is designed to provide mechanical support, control the water vapor permeability and to elute the antibiotic drug cloxacillin and the analgesic drug bupivacaine to the wound site. The porous sub-layer is designed to absorb the wound exudates and release the hemostatic agent tranexamic acid for bleeding control. The results show that the formulation parameters, i.e. crosslinker and plasticizer concentrations, affected the mechanical properties of the wound dressings as well as relevant physical properties (water vapor transmission rate and swelling kinetics), but had almost no effect on the drug-release profiles. While the antibiotic drug and the analgesic drug were released within several hours, the hemostatic agent was released within several minutes, according to the well designed hybrid structure. In conclusion, our novel soy protein hybrid wound dressings are biocompatible, can deliver various drugs simultaneously in a controlled fashion for each drug individually, and can be adjusted to suit various types of wounds by altering their properties through formulation effects.


Subject(s)
Bandages , Soybean Proteins/chemistry , Wound Healing , Analgesics/chemistry , Analgesics/pharmacokinetics , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Biocompatible Materials/chemistry , Cell Line , Cell Survival/drug effects , Delayed-Action Preparations , Hemostatics/chemistry , Hemostatics/pharmacokinetics , Humans , Materials Testing , Polymers/chemistry , Polymers/pharmacokinetics , Porosity , Wound Healing/drug effects
7.
Thromb Haemost ; 120(10): 1407-1416, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32746466

ABSTRACT

OBJECTIVE: Most von Willebrand disease (VWD) patients can be treated with desmopressin during bleeding or surgery. Large interpatient variability is observed in von Willebrand factor (VWF) activity levels after desmopressin administration. The aim of this study was to develop a pharmacokinetic (PK) model to describe, quantify, and explain this variability. METHODS: Patients with either VWD or low VWF, receiving an intravenous desmopressin test dose of 0.3 µg kg-1, were included. A PK model was derived on the basis of the individual time profiles of VWF activity. Since no VWF was administered, the VWF dose was arbitrarily set to unity. Interpatient variability in bioavailability (F), volume of distribution (V), and clearance (Cl) was estimated. RESULTS: The PK model was developed using 951 VWF activity level measurements from 207 patients diagnosed with a VWD type. Median age was 28 years (range: 5-76), median predose VWF activity was 0.37 IU/mL (range: 0.06-1.13), and median VWF activity response at peak level was 0.64 IU/mL (range: 0.04-4.04). The observed PK profiles were best described using a one-compartment model with allometric scaling. While F increased with age, Cl was dependent on VWD type and sex. Inclusion resulted in a drop in interpatient variability in F and Cl of 81.7 to 60.5% and 92.8 to 76.5%, respectively. CONCLUSION: A PK model was developed, describing VWF activity versus time profile after desmopressin administration in patients with VWD or low VWF. Interpatient variability in response was quantified and partially explained. This model is a starting point toward more accurate prediction of desmopressin dosing effects in VWD.


Subject(s)
Deamino Arginine Vasopressin/pharmacokinetics , Hemostatics/pharmacokinetics , von Willebrand Diseases/drug therapy , von Willebrand Factor/metabolism , Adolescent , Adult , Aged , Child , Child, Preschool , Deamino Arginine Vasopressin/therapeutic use , Female , Hemostatics/therapeutic use , Humans , Male , Middle Aged , Models, Biological , Young Adult , von Willebrand Diseases/metabolism
8.
Thromb Haemost ; 120(7): 1056-1065, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32480417

ABSTRACT

BACKGROUND: von Willebrand factor (VWF) is crucial for optimal dosing of factor VIII (FVIII) concentrate in hemophilia A patients as it protects FVIII from premature clearance. To date, it is unknown how VWF behaves and what its impact is on FVIII clearance in the perioperative setting. AIM: To investigate VWF kinetics (VWF antigen [VWF:Ag]), VWF glycoprotein Ib binding (VWF:GPIbM), and VWF propeptide (VWFpp) in severe and moderate perioperative hemophilia A patients included in the randomized controlled perioperative OPTI-CLOT trial. METHODS: Linear mixed effects modeling was applied to analyze VWF kinetics. One-way and two-way analyses of variance were used to investigate perioperative VWFpp/VWF:Ag ratios and associations with surgical bleeding. RESULTS: Fifty-nine patients with median age of 48.8 years (interquartile range: 34.8-60.0) were included. VWF:Ag and VWF:GPIbM increased significantly postoperatively. Blood type non-O or medium risk surgery were associated with higher VWF:Ag and VWF:GPIbM levels compared with blood type O and low risk surgery. VWFpp/VWF:Ag was significantly higher immediately after surgery than 32 to 57 hours after surgery (p < 0.001). Lowest VWF:Ag quartile (0.43-0.92 IU/mL) was associated with an increase of FVIII concentrate clearance of 26 mL/h (95% confidence interval: 2-50 mL/h) compared with highest VWF antigen quartile (1.70-3.84 IU/mL). VWF levels were not associated with perioperative bleeding F(4,227) = 0.54, p = 0.710. CONCLUSION: VWF:Ag and VWF:GPIbM levels increase postoperatively, most significantly in patients with blood type non-O or medium risk surgery. Lower VWF antigen levels did not lead to clinically relevant higher FVIII clearance. VWF:Ag or VWF:GPIbM levels were not associated with perioperative hemorrhage.


Subject(s)
Blood Loss, Surgical/prevention & control , Factor VIII/pharmacokinetics , Hemophilia A/drug therapy , Hemostatics/pharmacokinetics , Postoperative Hemorrhage/prevention & control , von Willebrand Factor/metabolism , Adult , Drug Administration Schedule , Drug Dosage Calculations , Factor VIII/administration & dosage , Factor VIII/adverse effects , Hemophilia A/blood , Hemophilia A/diagnosis , Hemostatics/administration & dosage , Hemostatics/adverse effects , Humans , Male , Metabolic Clearance Rate , Middle Aged , Netherlands , Perioperative Care , Treatment Outcome
9.
Thromb Haemost ; 120(5): 728-736, 2020 May.
Article in English | MEDLINE | ID: mdl-32369844

ABSTRACT

BACKGROUND: Factor VIII (FVIII) trough levels > 1 IU/dL in patients with severe hemophilia A receiving regular prophylaxis may optimize bleed protection. OBJECTIVES: In this post hoc analysis of patients receiving tertiary prophylaxis for approximately 1 year, the relationship between estimated FVIII levels and reported bleeds was investigated to predict the potential for zero bleeds. METHODS: Sixty-three patients (median [range] age, 28 [7-59] years) with severe hemophilia A (229 bleeds) were included. FVIII levels at time of each bleed were estimated from single-dose individual pharmacokinetics. The highest estimated FVIII level at which patients experienced a bleed was considered the "potentially effective trough level" for that bleed type. Kaplan-Meier estimates of proportions of patients with no bleeds above certain estimated FVIII levels were determined. Those not experiencing a bleed in the trial were assumed to have a bleed at 0 IU/dL (pragmatic approach) or at their median trough level (conservative approach). RESULTS: Kaplan-Meier estimates based on pragmatic approach predicted zero all bleeds, joint bleeds, and spontaneous joint bleeds in 1 year in 40, 43, and 63% of patients, respectively, when the potentially effective trough FVIII level was set at 1 IU/dL. Between 1 and 10 IU/dL, every 1 IU/dL rise in estimated FVIII level was associated with an additional 2% of patients having zero all bleeds. CONCLUSION: This post hoc analysis confirms benefits with trough levels of approximately 1 to 3 IU/dL in most patients starting tertiary prophylaxis; prophylaxis with higher trough levels may help patients to achieve zero bleeds.


Subject(s)
Factor VIII/pharmacokinetics , Hemarthrosis/prevention & control , Hemophilia A/drug therapy , Hemostasis/drug effects , Hemostatics/pharmacokinetics , Models, Biological , Tertiary Prevention , Adolescent , Adult , Canada , Child , Europe , Factor VIII/administration & dosage , Hemarthrosis/blood , Hemarthrosis/diagnosis , Hemophilia A/blood , Hemophilia A/diagnosis , Hemostatics/administration & dosage , Hemostatics/blood , Humans , Middle Aged , Risk Factors , Severity of Illness Index , United States , Young Adult
10.
Thromb Haemost ; 120(5): 737-746, 2020 May.
Article in English | MEDLINE | ID: mdl-32369845

ABSTRACT

Long-term safety and efficacy data of extended half-life factor IX (FIX) prophylaxis in children with hemophilia B (HB) are sparse. paradigm 5 is a multinational, open-label, single-arm, phase III trial assessing once-weekly (40 IU/kg) prophylactic nonacog beta pegol (N9-GP) in previously treated patients (PTPs) aged ≤ 12 years with HB (FIX activity ≤ 2%). Primary endpoint: incidence of anti-FIX inhibitory antibodies (≥ 0.6 Bethesda Units). We present a 5-year analysis (N = 25, including remaining patients with ≥ 5 years' follow-up) and compare with a 1-year analysis (≥ 52 weeks' exposure). The main phase enrolled 25 children; 22 entered the extension phase; 17 remained in trial at data cutoff. Median treatment period: 5.6 years/patient; median total number of N9-GP exposure days: 290.0/patient. No patients developed anti-FIX inhibitory antibodies. No other safety concerns, including thromboembolic events, were reported. Neurological examinations have not revealed any new abnormal findings. Sixteen (64.0%) patients remained free from spontaneous bleeds; all bleeds were mild/moderate in severity; 93.0% were controlled with 1 to 2 N9-GP injections. No intracranial hemorrhages were reported. Annualized bleeding rates (ABRs) were very low at 5 years (median/Poisson-estimated mean overall ABR: 0.66/0.99), having decreased from the 1-year analysis (1.00/1.44). Median/Poisson-estimated mean spontaneous ABRs for the 1- and 5-year analyses: 0.00/0.45 and 0.00/0.33. Mean FIX trough activity at 5 years: 17.9%. Mean polyethylene glycol plasma concentration reached steady state at 6 months, increasing slightly over time, in line with increased FIX trough activity. N9-GP administered for ≥ 5 years shows favorable long-term safety and efficacy in PTPs with HB (FIX activity ≤ 2%).


Subject(s)
Factor IX/administration & dosage , Hemophilia B/drug therapy , Hemostatics/administration & dosage , Polyethylene Glycols/administration & dosage , Adolescent , Age Factors , Asia , Child , Child, Preschool , Drug Administration Schedule , Europe , Factor IX/adverse effects , Factor IX/pharmacokinetics , Hemophilia B/blood , Hemophilia B/diagnosis , Hemorrhage/blood , Hemorrhage/diagnosis , Hemorrhage/prevention & control , Hemostatics/adverse effects , Hemostatics/blood , Hemostatics/pharmacokinetics , Humans , Infant , North America , Patient Safety , Polyethylene Glycols/adverse effects , Polyethylene Glycols/pharmacokinetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome
11.
Thromb Haemost ; 120(5): 747-757, 2020 May.
Article in English | MEDLINE | ID: mdl-32369846

ABSTRACT

BACKGROUND: The currently published population pharmacokinetic (PK) models used for PK-guided dosing in hemophilia patients are based on clinical trial data and usually not externally validated in clinical practice. The aim of this study was to validate a published model for recombinant factor VIII-Fc fusion protein (rFVIII-Fc) concentrate and to develop an enriched model using independently collected clinical data if required. METHODS: Clinical data from hemophilia A patients treated with rFVIII-Fc concentrate (Elocta) participating in the United Kingdom Extended Half-Life Outcomes Registry were collected. The predictive performance of the published model was assessed using mean percentage error (bias) and mean absolute percentage error (inaccuracy). An extended population PK model was developed using nonlinear mixed-effects modeling (NONMEM). RESULTS: A total of 43 hemophilia A patients (FVIII ≤ 2 IU/dL), aged 5 to 70 years, were included. The prior model was able to predict the collected 244 rFVIII-Fc levels without significant bias (-1.0%, 95% CI: -9.4 to 7.3%) and with acceptable accuracy (12.9%). However, clearance and central distribution volume were under predicted in patients <12 years, which was expected as this age group was not represented in the previous model population. An enriched population PK model was constructed, which was able to successfully characterize PK profiles of younger children. CONCLUSION: We concluded that the existing rFVIII-Fc population PK model is valid for patients ≥ 12 years. However, it is not reliable in younger patients. Our alternative model, constructed from real world patient data including children, allows for better description of patients ≥5 years.


Subject(s)
Factor VIII/pharmacokinetics , Hemophilia A/drug therapy , Hemostatics/pharmacokinetics , Models, Biological , Recombinant Fusion Proteins/pharmacokinetics , Adolescent , Adult , Age Factors , Aged , Child , Factor VIII/administration & dosage , Factor VIII/adverse effects , Hemophilia A/blood , Hemophilia A/diagnosis , Hemostatics/administration & dosage , Hemostatics/adverse effects , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/adverse effects , Male , Middle Aged , Netherlands , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , Registries , Reproducibility of Results , United Kingdom , Young Adult
12.
Invest New Drugs ; 38(5): 1580-1587, 2020 10.
Article in English | MEDLINE | ID: mdl-32166534

ABSTRACT

Purpose The vasopressin analog desmopressin (dDAVP) is known to increase plasma levels of hemostatic factors, and preclinical studies in colorectal cancer models have demonstrated that it hampers tumor vascularization and metastatic progression. We evaluated safety and preliminary efficacy of dDAVP in rectal cancer patients with bleeding, before receiving specific oncologic treatment with surgery, chemotherapy and/or radiotherapy. Methods Patients with rectal cancer having moderate or severe rectal bleeding were enrolled in an open-label, dose-finding trial. Intravenous infusions of dDAVP were administered during two consecutive days in doses from 0.25 to 2.0 µg/kg, using single or twice daily regimen. Bleeding was graded using a score based on the Chutkan scale and tumor perfusion was evaluated by dynamic contrast-enhanced magnetic resonance imaging. Results The trial accrued a total of 32 patients. Dose-limiting toxicity occurred in patients receiving 1 µg/kg or higher. The most prominent treatment-related severe adverse event was hyponatremia. Most patients receiving the maximum tolerated dose of 0.5 µg/kg showed at least a partial hemostatic response and 58% developed a complete response with absence of bleeding at day 4 and/or at the last follow-up at day 14. Tumor perfusion was decreased in two-thirds of patients after dDAVP treatment. Conclusions dDAVP appeared as a promising hemostatic agent in rectal cancer patients with bleeding. Randomized clinical trials to confirm its effectiveness are warranted.Clinical trial registration www.clinicaltrials.gov NCT01623206.


Subject(s)
Deamino Arginine Vasopressin/administration & dosage , Hemorrhage/drug therapy , Hemostatics/administration & dosage , Rectal Neoplasms/drug therapy , Adult , Aged , Deamino Arginine Vasopressin/adverse effects , Deamino Arginine Vasopressin/pharmacokinetics , Hemorrhage/metabolism , Hemostatics/adverse effects , Hemostatics/pharmacokinetics , Humans , Infusions, Intravenous , Male , Middle Aged , Rectal Neoplasms/metabolism , Treatment Outcome , Young Adult
13.
J Thromb Haemost ; 17(8): 1240-1246, 2019 08.
Article in English | MEDLINE | ID: mdl-31148392

ABSTRACT

BACKGROUND: Following induced joint hemorrhage, hemophilia B results in the abnormal persistence of iron deposition, inflammation, and neovascularity of the synovial tissue, as well as deterioration of the bone articular surface and strength. Previously, we demonstrated that a factor IX (FIX) replacement protein with extended circulating FIX activity, glycoPEGylated FIX nonacog beta pegol (N9-GP), could improve synovial and osteochondral parameters in F9 knockout mice when administered after joint injury. OBJECTIVE: We explored the use of N9-GP prior to unilateral joint hemorrhage and compared to unmodified recombinant FIX (rFIX). METHODS: Pharmacodynamics, histology, and microcomputed tomography were used to assess the effects of prophylactic administration of glycoPEGylated FIX. RESULTS: In comparison to rFIX, N9-GP significantly improved soft tissue histological parameters, as well as bone outcome at 2 weeks post injury, while performing equally in reduction of blood present in the joint space assessed 1 day after injury. CONCLUSIONS: These results indicate that, in comparison to rFIX, the prophylactic use of extended half-life FIX provides superior protection from bleeding-induced joint damage, manifested by improved correction of histologic parameters.


Subject(s)
Factor IX/metabolism , Hemarthrosis/drug therapy , Hemophilia B/drug therapy , Hemostatics/administration & dosage , Joints/drug effects , Polyethylene Glycols/administration & dosage , Animals , Disease Models, Animal , Drug Administration Schedule , Factor IX/administration & dosage , Factor IX/genetics , Factor IX/pharmacokinetics , Half-Life , Hemarthrosis/diagnostic imaging , Hemarthrosis/genetics , Hemarthrosis/metabolism , Hemophilia B/genetics , Hemophilia B/metabolism , Hemostatics/pharmacokinetics , Joints/diagnostic imaging , Joints/pathology , Mice, Inbred C57BL , Mice, Knockout , Polyethylene Glycols/pharmacokinetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics
14.
J Thromb Haemost ; 17(8): 1288-1296, 2019 08.
Article in English | MEDLINE | ID: mdl-31055871

ABSTRACT

BACKGROUND: Optimization of factor VIII (FVIII) infusion in hemophilia A would benefit from identification of FVIII pharmacokinetics (PK) determinants. The low-density lipoprotein receptor (LDLR) contains an FVIII-binding site and might influence FVIII clearance. Consistently, LDLR polymorphisms have been associated with FVIII levels. OBJECTIVE: To investigate the relationships between individual FVIII PK and functional LDLR polymorphisms. PATIENTS/METHODS: Thirty-three hemophilia A patients (FVIII coagulant activity [FVIII:C] ≤2 IU/dL) without inhibitors underwent 85 FVIII single-dose (21.4-51.8 IU/kg) PKs with different FVIII concentrates. Twenty patients underwent repeated PKs (2-6). FVIII: C measured up to 72 hours was analyzed by two-compartment model. Parameters were evaluated in relation to F8 mutations, ABO blood-group and LDLR genotypes. RESULTS: F8 mutation types were not associated with PK parameters. ABO and LDLR c.1773C/T polymorphism were associated with Alpha, Alpha HL, CLD2, K1-2, and K2-1 parameters, suggesting an influence on the FVIII initial distribution phase. Regression analysis showed an independent association of both ABO and LDLR c.1773C/T with PK parameters (Alpha, ß-coefficient -0.311 vs 0.348; CLD2, ß-coefficient -0.335 vs 0.318), giving rise to an additive effect in subjects stratified by combined phenotypes. Differently, the LDLR c.81C/T was associated with FVIII clearance and volume of distribution at steady state, which could be related to distinct effects of polymorphisms, potentially linked to LDLR intracellular distribution and FVIII binding behavior. CONCLUSIONS: With the limitation of different FVIII concentrates and low number of patients, our data show plausible associations of LDLR polymorphisms with FVIII PK parameters, thus supporting their investigation as candidate functional determinants of FVIII PK.


Subject(s)
Factor VIII/pharmacokinetics , Hemophilia A/drug therapy , Hemostatics/pharmacokinetics , Pharmacogenomic Variants , Polymorphism, Genetic , Receptors, LDL/genetics , ABO Blood-Group System , Adolescent , Adult , Aged , Drug Monitoring , Factor VIII/administration & dosage , Factor VIII/genetics , Hemophilia A/blood , Hemophilia A/diagnosis , Hemophilia A/genetics , Hemostatics/administration & dosage , Humans , Metabolic Clearance Rate , Middle Aged , Models, Biological , Young Adult
15.
J Thromb Haemost ; 17(7): 1085-1096, 2019 07.
Article in English | MEDLINE | ID: mdl-31038793

ABSTRACT

Essentials The PK parameters of Eloctate vs Adynovate were compared using one-stage and chromogenic assays in 25 boys (12-18 years). The FVIII levels were taken at 3, 24, 48, and 72 hours following a dose of either FVIII; levels analyzed by WAPPS PK program. The PK profiles (half-life, clearance, and time to 5%, 3%, and 1%) were not statistically different for the two EHL FVIIIs. The significant interpatient variability in PK is mainly related to VWF levels (and blood group). BACKGROUND: A head-to-head comparison of the pharmokinetcs (PK) of extended half-life (EHL) factor VIII (FVIII) concentrates in the same subjects has not been reported. Recently, boys (ages 12-18 years) with hemophilia A in Canada were required to switch from Eloctate to Adynovate. OBJECTIVES: Compare the PK profiles of Eloctate vs Adynovate in the same boys. METHODS: Boys switching from Eloctate to Adynovate prophylaxis had FVIII levels sampled at 3, 24, 48, and 72 hours following a regular prophylactic infusion of Eloctate and then 1-3 months later, of Adynovate. Testing was done by one-stage assay (OSA) and chromogenic assay (CA). The PK parameters were determined with the Web Accessible Population Pharmacokinetic Service (WAPPS)-Hemo PK tool. RESULTS: Twenty-five boys (mean age 15.3 years; range: 12.1-18.4; 9 O blood group) underwent switching. Mean (range) terminal half-lives with the OSA were 16.1 hours (10.4 to 23.4; Eloctate) and 16.7 hours (11.0 to 23.6; Adynovate) (NS). With the CA, these were 18.0 hours (12.0 to 25.5; Eloctate) and 16.0 hours (10.3 to 22.9; Adynovate) (P = 0.001). There were no significant differences between the two EHL-FVIIIs in clearance, area under the concentration vs time curve (AUC), Vss, or time for FVIII levels to drop to 5%, 3%, and 1%. At the 72-h time point, mean observed FVIII levels following a mean dose of 39.3 IU/kg of Eloctate were 4.4% (OSA) and 4.4% (CA). For Adynovate, these were 5.1% (OSA) and 5.3% (CA) following similar doses. There was considerable interpatient variation in PK, mainly explained by differences in blood group/von Willebrand factor (VWF) levels. CONCLUSIONS: Eloctate and Adynovate have almost identical PK parameters. When switching from one to another no prophylaxis regimen change is needed.


Subject(s)
Factor VIII/pharmacokinetics , Hemophilia A/drug therapy , Hemostatics/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , ABO Blood-Group System , Adolescent , Child , Drug Substitution , Factor VIII/administration & dosage , Half-Life , Hemophilia A/blood , Hemophilia A/diagnosis , Hemostatics/administration & dosage , Humans , Immunoglobulin Fc Fragments/administration & dosage , Male , Metabolic Clearance Rate , Ontario , Recombinant Fusion Proteins/administration & dosage , Severity of Illness Index , von Willebrand Factor/metabolism
16.
Int J Lab Hematol ; 41(3): 325-330, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30735311

ABSTRACT

INTRODUCTION: Patients with Type 1 von Willebrand disease (VWD) have reduced amounts of von Willebrand factor (VWF) in their blood. Desmopressin (DDAVP) has been used to raise the blood levels of VWF in these patients. However, not all patients with Type 1 VWD are known to respond to DDAVP therapy. We sought to compare the levels of Factor VIII, VWF antigen (VWF:Ag), VWF ristocetin cofactor (VWF:RCo), and fibrinogen at different time points after administration of DDAVP. METHODS: A retrospective analysis was conducted at a single institution on 89 patients with Type 1 VWD who received a single intravenous dose of 0.3 µg/kg DDAVP. Levels of Factor VIII, VWF:Ag, VWF:RCo, and fibrinogen were measured before and then 30, 60, 90, and 120 minutes after completion of DDAVP administration. RESULTS: Median levels of Factor VIII, VWF:Ag, and VWF:RCo were significantly elevated (P < 0.001) at 30 (175 IU/dL, 111 IU/dL, 127 IU/dL), 60 (218 IU/dL, 138 IU/dL, 139 IU/dL), and 90 (212 IU/dL, 134 IU/dL, 130 IU/dL) minutes time points when compared to their respective baseline values (71 IU/dL, 54 IU/dL, 54 IU/dL). At 60 minutes, 96% of subjects were responders, but three subjects failed to respond at any time point. CONCLUSION: Measurement of levels of Factor VIII, VWF:Ag, and VWF:RCo, optimally at 60 minutes after the DDAVP infusion, is sufficient to confirm a patient as a responder in a DDAVP challenge test. Curtailing the number of time points of measurement will result in significant savings in cost and time to patients and their providers.


Subject(s)
Deamino Arginine Vasopressin/therapeutic use , von Willebrand Disease, Type 1/blood , von Willebrand Disease, Type 1/drug therapy , Adult , Biomarkers , Blood Coagulation , Blood Coagulation Factors , Blood Coagulation Tests/methods , Deamino Arginine Vasopressin/administration & dosage , Female , Hemostatics/pharmacokinetics , Hemostatics/therapeutic use , Humans , Male , Middle Aged , Retrospective Studies , Time Factors , Treatment Outcome , von Willebrand Disease, Type 1/diagnosis , von Willebrand Factor
17.
J Thromb Haemost ; 17(3): 460-469, 2019 03.
Article in English | MEDLINE | ID: mdl-30614620

ABSTRACT

Essentials Hemophilia patients on concizumab prophylaxis may need rFVIIa to treat breakthrough bleeds. Effect and safety of concizumab + rFVIIa were tested in vitro and in vivo. Concizumab + rFVIIa had no additive effects on bleeding in hemophilic rabbits. High steady-state levels of concizumab did not affect the safety of rFVIIa in cynomolgus monkeys. SUMMARY: Background Concizumab is a monoclonal antibody (mAb) against tissue factor pathway inhibitor (TFPI), currently in clinical development as a subcutaneous prophylactic therapy for hemophilia A/B with and without inhibitors. In patients with inhibitors, the treatment choice for breakthrough bleeding will comprise bypassing agents, e.g. activated recombinant FVIIa (rFVIIa) or activated prothrombin complex concentrates. Objectives To explore the effect and safety of concizumab and rFVIIa when they are simultaneously present. Methods Human blood made hemophilic with a FVIII antibody was spiked with increasing concentrations of concizumab, rFVIIa, or concizumab and rFVIIa in combination, and this was followed by thrombin generation test or thromboelastography. Blood loss in hemophilic rabbits was measured when concizumab, rFVIIa or concizumab + rFVIIa was administered either before or during cuticle bleeding. In a safety study, cynomolgus monkeys were exposed to high steady-state concizumab concentrations and given three doses of rFVIIa, and then subjected to full necropsy and histopathological examination. Results In human blood, concizumab + rFVIIa had more pronounced procoagulant effects under hemophilic conditions than the sum of individual responses. In contrast, concizumab + rFVIIa had no additional effects on blood loss in hemophilic rabbits as compared with rFVIIa or concizumab alone. In cynomolgus monkeys, the macroscopic and microscopic pathological examinations revealed no thrombi or other signs of excessive coagulation activation. Both rFVIIa and concizumab caused increases in thrombin-antithrombin and D-dimer concentrations; this effect tended to be additive with concomitant administration. Conclusions Concizumab did not affect the potency or safety of rFVIIa in vivo. These results support a clinical evaluation of rFVIIa at standard dose (90 µg kg-1 ) to treat breakthrough bleeds in concizumab clinical trials.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Factor VIIa/administration & dosage , Hemophilia A/drug therapy , Hemorrhage/drug therapy , Hemostasis/drug effects , Hemostatics/administration & dosage , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/toxicity , Disease Models, Animal , Drug Interactions , Factor VIIa/pharmacokinetics , Factor VIIa/toxicity , Female , Hemophilia A/blood , Hemorrhage/blood , Hemostatics/pharmacokinetics , Hemostatics/toxicity , Humans , Macaca fascicularis , Rabbits , Toxicokinetics
19.
J Thromb Haemost ; 17(4): 635-644, 2019 04.
Article in English | MEDLINE | ID: mdl-30661302

ABSTRACT

Essentials A novel fibrinogen concentrate was evaluated in patients with congenital fibrinogen deficiency. An open-label, phase 2-3 trial studied pharmacology, efficacy, and safety in patients >6 years. The product offers safe and effective therapy in the treatment and prophylaxis of bleeding. Data in recovery show the need of adjusted treatment and further investigation in children. SUMMARY: Background Single-factor replacement therapy is considered the most suitable treatment option for hereditary fibrinogen deficiency. A triple-secured plasma-derived human fibrinogen product was developed to increase the safety of the former fibrinogen concentrate. Objectives This non-randomized, open-label, prospective study investigated pharmacokinetics, efficacy, and safety of a novel fibrinogen concentrate (FibCLOT® /CLOTTAFACT® LFB, France) in inherited deficiency. Patients/Methods Fourteen patients ≥40 kg received fibrinogen concentrate for pharmacology and 16 ≥ 23 kg received treatment for bleeding or surgery. Each treatment was followed by a 3-week safety observation period. Key outcomes included number of infusions, dose, bleeding control, daily assessment, hemoglobin, blood loss, transfusions, and physicians' global assessment of response. Results Incremental recovery was 2.35 mg mL-1  per mg kg-1 and maximal concentration 1.41 g L-1 (geometric mean) after 0.060 g kg-1 infusion in 14 afibrinogenemic patients. Terminal half-life was 69.3 h (non-compartmental analysis). The maximum clot firmness was increased by a mean of 10.3 mm from baseline to maximal effect. Sixteen patients participated to the efficacy phase: 32 bleeding episodes were treated in 9 patients, and 15 patients underwent 38 surgical/invasive procedures. All patients achieved appropriate hemostasis: response to treatment was successful in all bleeds (95% CI, 0.89-1.00) and procedures (95% CI, 0.91-1.00). Most (94%) bleeds were controlled with a single infusion (median 0.050 g kg-1 ). Two patients experienced asymptomatic distal venous thromboses identified by systematic ultrasound. Conclusion FibCLOT® /CLOTTAFACT® showed a pharmacokinetic profile comparable to that of other fibrinogen concentrates and provides safe and clinically effective substitution therapy for fibrinogen-deficient patients.


Subject(s)
Afibrinogenemia/drug therapy , Fibrinogen/administration & dosage , Hemorrhage/drug therapy , Hemostasis/drug effects , Hemostatics/administration & dosage , Adolescent , Adult , Afibrinogenemia/blood , Afibrinogenemia/congenital , Afibrinogenemia/diagnosis , Age Factors , Child , Female , Fibrinogen/adverse effects , Fibrinogen/pharmacokinetics , Hemorrhage/blood , Hemorrhage/congenital , Hemorrhage/diagnosis , Hemostatics/adverse effects , Hemostatics/pharmacokinetics , Humans , Male , Prospective Studies , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL