Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Haemophilia ; 27(6): 1007-1021, 2021 Nov.
Article En | MEDLINE | ID: mdl-34494337

INTRODUCTION: Type 2N von Willebrand disease (VWD) is characterized by a decreased affinity of von Willebrand factor (VWF) for factor VIII (FVIII). Abnormal binding of FVIII to VWF (VWF:FVIIIB), results in low FVIII plasma levels, which can lead to a misdiagnosis of mild haemophilia A. Accurate diagnosis of type 2N VWD is essential for appropriate genetic counselling and therapy. This disease can be distinguished from haemophilia A by in vitro assays (measurement VWF:FVIIIB activity) and/or genetic analysis. AIM: To identify the current challenges in the diagnosis and treatment of this type of VWD and provide an in-depth description of the phenotypes and mutations identified. RESULTS: Twenty-eight patients had at least one type 2N mutation, and 13 of these had a type 2N mutation combined with other variations. Three type 2N mutations were detected: p.Arg816Trp, p.Arg854Gln, and p.Arg763Ser. Two of these are the most frequently described mutations worldwide. This mutational spectrum differs from the broad spectrum seen in neighbouring France, where at least eight distinct 2N mutations have been found. In the PCM-EVW-ES cohort, 11 asymptomatic type 2N carriers with borderline FVIII plasma levels would probably have been excluded if the evaluation had been based on clinical and laboratory data only. Likewise, three patients with a severe phenotype would have been classified as homozygous for a 2N mutation if only the phenotype study had been performed. CONCLUSION: The high detection yield and affordability of next-generation sequencing support the use of this technology as a first-line diagnostic tool in this setting.


Hemophilia A , von Willebrand Disease, Type 2 , von Willebrand Diseases , von Willebrand Factor/genetics , Factor VIII/genetics , Heterozygote , Homozygote , Humans , von Willebrand Disease, Type 2/diagnosis , von Willebrand Disease, Type 2/genetics
2.
Thromb Haemost ; 121(11): 1400-1408, 2021 Nov.
Article En | MEDLINE | ID: mdl-33581698

INTRODUCTION: FVIII inhibitor development is the most serious contemporary treatment complication in haemophilia A, particularly in previously untreated patients (PUPs). No inhibitors developed in clinical trials in previously treated patients treated with simoctocog alfa (Nuwiq), a fourth-generation recombinant FVIII produced in a human cell line. METHODS: The NuProtect study investigated the immunogenicity of simoctocog alfa in PUPs. NuProtect was a prospective, multinational, open-label, non-controlled, phase III study. PUPs with severe haemophilia A (FVIII:C <1%) of any age and ethnicity were treated with simoctocog alfa for 100 exposure days or a maximum of 5 years. Patients were true PUPs without prior exposure to FVIII concentrates or blood components. Inhibitor titres were measured with the Nijmegen-modified Bethesda assay; cut-off for positivity was 0.6 BU mL-1 (≥0.6 to <5 low-titre, ≥5 high titre). RESULTS: A total of 108 PUPs with a median age at first treatment of 12.0 months (interquartile range: 8.0-23.5) were treated with simoctocog alfa. F8 mutation type was known for 102 patients (94.4%) of whom 90 (88.2%) had null F8 mutations and 12 (11.8%) had non-null mutations. Of 105 PUPs evaluable for inhibitor development, 28 (26.7%) developed inhibitors; 17 high titre (16.2%) and 11 low titre (10.5%). No PUPs with non-null F8 mutations developed inhibitors. CONCLUSION: In the NuProtect study, the rate of inhibitor development in PUPs with severe haemophilia A treated with simoctocog alfa was lower than the rate reported for hamster-cell-derived recombinant factor VIII products in other recent clinical trials. No inhibitors were reported in PUPs with non-null F8 mutations.


Antibodies/blood , Coagulants/therapeutic use , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemorrhage/prevention & control , Coagulants/immunology , Factor VIII/genetics , Factor VIII/immunology , Genetic Predisposition to Disease , Hemophilia A/blood , Hemophilia A/genetics , Hemorrhage/blood , Hemorrhage/diagnosis , Hemorrhage/genetics , Humans , Infant , Male , Mutation , Prospective Studies , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Severity of Illness Index , Time Factors , Treatment Outcome
3.
Haemophilia ; 26(4): 637-642, 2020 Jul.
Article En | MEDLINE | ID: mdl-32542961

INTRODUCTION: Adherence to prophylaxis regimens is essential for bleed prevention in haemophilia but remains a challenge due to the need for frequent infusions. AIM: To evaluate patient adherence to prophylaxis regimens with a long-acting recombinant factor IX (rIX-FP; IDELVION® ) in clinical studies and real-world practice. METHODS: In two phase 3 clinical studies, patients with haemophilia B (FIX ≤2%) recorded their dose, dosing frequency and rIX-FP consumption in an e-diary. Adherence to prescribed prophylaxis regimens was assessed in all patients and to prescribed dose in patients ≥12 years only. Additionally, adherence to rIX-FP prophylaxis regimens in real-world practice was captured. RESULTS: In clinical studies, 94.9% (n = 56/59) of patients ≥12 years and 100% (n = 27) of paediatric patients received ≥80% of the expected number of infusions for their assigned prophylaxis schedule. Overall, mean adherence rate was 95.5% across all prophylaxis regimens in patients ≥12 years and 97.9% with a 7-day regimen in paediatric patients. In patients ≥12 years, 85.7% (n = 54/63) were dose adherent, defined as receiving within 10% of their prescribed dose ≥80% of the time. In real-world practice, adherence was observed in 100% (n = 14 and n = 15, respectively) of patients in two haemophilia treatment centres and 57.1% (n = 4/7) of patients in a third centre; non-adherence (n = 3/7) was linked to insurance-related and parental issues. CONCLUSION: In clinical studies, patients with haemophilia B had high adherence rates to rIX-FP prophylaxis regimens with a variety of dosing intervals, enabling them to achieve very low bleeding rates. High adherence may also be achievable in real-world practice.


Factor IX/therapeutic use , Hemophilia B/drug therapy , Hemorrhage/prevention & control , Recombinant Fusion Proteins/therapeutic use , Serum Albumin/therapeutic use , Treatment Adherence and Compliance/statistics & numerical data , Adolescent , Adult , Aged , Child , Factor IX/administration & dosage , Hemophilia B/complications , Hemorrhage/etiology , Humans , Infusions, Intravenous , Middle Aged , Practice Patterns, Physicians' , Recombinant Fusion Proteins/administration & dosage , Serum Albumin/administration & dosage , Treatment Adherence and Compliance/psychology , Young Adult
4.
Res Pract Thromb Haemost ; 3(3): 397-404, 2019 Jul.
Article En | MEDLINE | ID: mdl-31294327

BACKGROUND: Prophylaxis reduces the frequency of bleeds in boys with severe hemophilia and is the standard care for their management in resource-abundant countries. The effect of prophylaxis on Health-Related Quality of Life (HRQoL) has not been established, because the sample sizes of most studies are too small to explore the relationship of multiple factors that influence HRQoL. METHODS: The aim of this study was to assess the impact of hemophilia severity and treatment regimen on HRQoL and to establish the minimum important difference (MID) using the international level of score distributions. HRQoL data were pooled from 7 studies across 9 countries. HRQoL was measured using the Canadian Hemophilia Outcomes-Kids' Life Assessment Tool (CHO-KLAT). A mixed-effect linear regression analysis was employed to assess the impact of prophylaxis on the CHO-KLAT score. RESULTS: Data from 401 boys with hemophilia were analyzed (57.6% severe hemophilia and 57.6% receiving prophylaxis). The model revealed that receiving prophylaxis was significantly associated with higher HRQoL (regression coefficient 8.5, 95% confidence interval [CI] 3.9-13.1). Boys with severe hemophilia had a significantly lower HRQoL as compared to boys with moderate and mild hemophilia whose CHO-KLAT scores were 7.0 and 6.6 points higher, respectively. There was a significant interaction between treatment and disease severity (P = 0.023), indicating prophylaxis has the most significant impact in boys with severe hemophilia. Based on these pooled data, the MID of the CHO-KLAT was established at 6.5. CONCLUSIONS: This study confirms the positive effect of prophylaxis on HRQoL in boys with hemophilia in a real-world setting and provides initial benchmarks for interpreting HRQoL scores based on use of the CHO-KLAT instrument.

6.
Haemophilia ; 25(1): 144-153, 2019 Jan.
Article En | MEDLINE | ID: mdl-30444298

AIM: The use of musculoskeletal ultrasound (MSK-US) following protocols for haemophilic arthropathy and the Haemophilia Early Arthropathy Detection with Ultrasound (HEAD-US) score can help standardize monitoring in haemophilia. This study evaluated the joint status (elbows, knees and ankles) of patients with haemophilia B (HB) in Spain using MSK-US and the HEAD-US score. METHODS: Haemophilia B patients ≥14 years old were included in this observational, multicentre, cross-sectional study, regardless of their clinical condition, HB severity and treatment received. Two blinded observers were involved in image acquisition and scoring in each centre. RESULTS: Eighty-two patients from 12 centres were enrolled: 27% mild HB, 23% moderate, 50% severe HB. Mean age was 38.9 ± 16.4 years, 60% were treated on demand (OD) and 40% were on prophylaxis. HEAD-US was zero in all joints in 28.6% OD patients and 36.4% on prophylaxis. Mean scores significantly worsened with HB severity, except for the left knee. Patients on primary and secondary prophylaxis had significantly better joint health vs OD patients in all joints, except the right ankle. Among OD patients, those with severe disease presented significantly worse scores in all HEAD-US items related to permanent damage. CONCLUSION: Joint status of HB patients in Spain is influenced by severity and treatment modality, related to the development of arthropathy, which appears prevalent in OD patients with severe HB. Routine assessment with an imaging tool such as ultrasound and HEAD-US system may help to improve joint health by personalizing and adjusting treatment in this population.


Hemophilia B/pathology , Joint Diseases/diagnosis , Joints/diagnostic imaging , Synovitis/diagnosis , Adolescent , Adult , Cross-Sectional Studies , Humans , Joint Diseases/pathology , Logistic Models , Middle Aged , Odds Ratio , Severity of Illness Index , Spain , Synovitis/pathology , Ultrasonography , Young Adult
7.
PLoS One ; 13(6): e0197876, 2018.
Article En | MEDLINE | ID: mdl-29924855

The multimeric analysis (MA) of plasma von Willebrand factor (VWF) evaluates structural integrity and helps in the diagnosis of von Willebrand disease (VWD). This assay is a matter of controversy, being considered by some investigators cumbersome and only slightly informative. The centralised study 'Molecular and Clinical Profile of von Willebrand Disease in Spain (PCM-EVW-ES)' has been carried out by including the phenotypic assessment and the genetic analysis by next generation sequencing (NGS) of the VWF gene (VWF). The aim of the present study was to evaluate the role of MA to the diagnosis of these patients and their potential discrepancies. Two hundred and seventy out of 480 patients centrally diagnosed with VWD had normal multimers, 168 had abnormal multimers and 42 a total absence of multimers. VWF MA was of great significance in the diagnosis of 83 patients (17.3%), it was also of help in the diagnosis achieved in 365 additional patients (76%) and was not informative in 32 cases (6.7%). With regard to discrepancies, 110 out of 480 (23%) patients centrally diagnosed with VWD presented some kind of discordance between VWF:RCo/VWF:Ag and/or VWF:CB/VWF:Ag ratios, multimeric study and/or genetic results. The VWF MA was key in the presence of novel mutations as well as in cases with phenotypic discrepancies. A comparison between the contribution of MA and VWF:CB showed a clearly higher contribution of the former in the diagnostic process. These data seem to reinforce the relevance of the VWF MA in VWD diagnosis, despite all its limitations.


High-Throughput Nucleotide Sequencing , von Willebrand Diseases/diagnosis , von Willebrand Diseases/genetics , von Willebrand Factor/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Genotype , Humans , Male , Middle Aged , Phenotype , Spain , Young Adult
8.
Thromb Haemost ; 118(3): 451-460, 2018 03.
Article En | MEDLINE | ID: mdl-29448295

Recombinant factor XIII-A2 (rFXIII-A2) was developed for prophylaxis and treatment of bleeds in patients with congenital FXIII A-subunit deficiency. mentor™2 (NCT00978380), a multinational, open-label, single-arm, multiple-dosing extension to the pivotal mentor™1 trial, assessed long-term safety and efficacy of rFXIII-A2 prophylaxis in eligible patients (patients with severe [<0.05 IU/mL] congenital FXIII subunit A deficiency) aged ≥6 years. Patients received 35 IU/kg rFXIII-A2 (exact dosing) every 28 ± 2 days for ≥52 weeks. Primary endpoint was safety (adverse events including immunogenicity); secondary endpoints were rate of bleeds requiring FXIII treatment, haemostatic response after one 35 IU/kg rFXIII-A2 dose for breakthrough bleeds and withdrawals due to lack of rFXIII-A2 efficacy. Steady-state pharmacokinetic variables were also summarized. Elective surgery was permitted during the treatment period. Sixty patients were exposed to rFXIII-A2; their median age was 26.0 years (range: 7.0-77.0). rFXIII-A2 was well tolerated without any safety concerns. No non-neutralizing or neutralizing antibodies (inhibitors) against FXIII were detected. Mean annualized bleeding rate (ABR) was 0.043/patient-year. Mean spontaneous ABR was 0.011/patient-year. No patients withdrew due to lack of efficacy. Geometric mean FXIII trough level was 0.17 IU/mL. Geometric terminal half-life was 13.7 days. rFXIII-A2 prophylaxis provided sufficient haemostatic coverage for 12 minor surgeries without the need for additional FXIII therapy; eight procedures were performed within 7 days of the patient's last scheduled rFXIII-A2 dose, and four were performed 10 to 21 days after the last dose.


Factor VIIIa/therapeutic use , Factor XIII Deficiency/therapy , Hemorrhage/prevention & control , Surgical Procedures, Operative , Adolescent , Adult , Aged , Child , Factor XIII Deficiency/congenital , Factor XIII Deficiency/surgery , Female , Hemostasis , Humans , Male , Middle Aged , Patient Safety , Recombinant Proteins/therapeutic use , Young Adult
9.
Thromb Haemost ; 117(12): 2274-2282, 2017 12.
Article En | MEDLINE | ID: mdl-29212115

In children with severe haemophilia A, inhibitors to factor VIII (FVIII) usually develop during the first 50 treatment exposure days and are classified as low or high titre depending on the peak inhibitor titre being greater or less than 5 Bethesda units/mL (BU/mL). Classification of the inhibitor may change with time, as some low-titre inhibitors progress to high titre following re-exposure to FVIII concentrate. The aim of this study was to investigate potential risk factors for such a progression in children with severe haemophilia A and newly diagnosed inhibitors. This study was a follow-up study of the PedNet Registry and included 260 children with severe haemophilia A and inhibitors born between 1990 and 2009 and recruited consecutively from 31 haemophilia centres. Clinical and laboratory data were collected from the date of each child's first positive inhibitor test for at least 3 years. At the time of first positive inhibitor test, 49% (n = 127) had low-titre inhibitors, with 50% of them progressing to high titre and only 25% maintaining low titres. The FVIII gene (F8) mutation type was known in 247 patients (95%), and included 202 (82%) null mutations. The progression to high-titre inhibitors was associated with null F8 mutations (odds ratio [OR]: 2.6; 95% confidence interval [CI]: 1.0­6.5), family history of inhibitors (OR: 7.2; 95% CI: 1.8­28.4) and the use of high-dose immune tolerance induction, defined as ≥100 IU FVIII concentrate/kg/d (OR: 3.9; 95% CI: 1.5­10.0). These results suggest that high-dose immune tolerance induction should be avoided as the initial strategy in patients who develop low-titre FVIII inhibitors.


Blood Coagulation Factor Inhibitors/therapeutic use , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Adolescent , Animals , Child , Child, Preschool , Disease Progression , Factor VIII/genetics , Female , Follow-Up Studies , Hemophilia A/epidemiology , Humans , Immune Tolerance , Male , Mutation/genetics , Risk Factors
10.
Clin Ther ; 38(4): 936-44, 2016 04.
Article En | MEDLINE | ID: mdl-26969334

PURPOSE: Nonacog alfa, a recombinant factor IX (FIX) product, is used for FIX replacement in the treatment and prevention of bleeding events in patients with hemophilia B. This study aimed to provide supplemental pharmacokinetic (PK), efficacy, and safety data for nonacog alfa when administered as part of usual hemophilia care, including on-demand treatment, routine prophylaxis, and surgical prophylaxis. METHODS: Men with previously treated severe or moderately severe hemophilia B (FIX activity ≤2%) were enrolled in this prospective, open-label, nonrandomized, multicenter study. An initial 72-hour PK assessment was performed wherein patients received a single dose of nonacog alfa (75 IU/kg) as an infusion over 10 minutes. A final 72-hour PK assessment was performed at the patient's last visit, after a minimum washout period of 4 days. Correlations between Cmax after the first dose and body weight and body mass index (BMI) were assessed post hoc using Spearman test after evaluating normality. FINDINGS: In total, 23 patients (age, 12-59 years; weight, 44-173 kg; and BMI, 16.3-45.1) with previous exposure to FIX products (median, 460 days; range, 150-2400 days) were enrolled; 21 were evaluable for efficacy. The median number of exposure days per efficacy-evaluable patient in this study was 48 (range, 31-103). The FIX activity profiles showed multiphasic disposition characteristics, with initial mean (SD) PK profiles as follows: Cmax, 61.4 (12.5) IU/dL; AUC∞, 1055 (227) IU·h/dL; t½, 23.7 (5.6) hours; and recovery, 0.818 (0.167) IU/dL. Mean plasma FIX activity versus time profiles were essentially identical upon initial exposure and after repeated use (n = 17), and bioequivalence was confirmed. No apparent relationship was observed between Cmax and either body weight (P > 0.1732) or BMI (P > 0.1235). IMPLICATIONS: The FIX activity profile after administration of nonacog alfa is predictable and is not altered after repeated exposure during usual hemophilia care. PK parameters are consistent with nonacog alfa use for FIX replacement in on-demand treatment, routine prophylaxis, and surgical prophylaxis in patients with hemophilia B.


Factor IX , Hemophilia B/drug therapy , Recombinant Proteins , Adolescent , Adult , Child , Factor IX/adverse effects , Factor IX/pharmacokinetics , Factor IX/therapeutic use , Humans , Male , Middle Aged , Prospective Studies , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Young Adult
11.
Blood ; 127(14): 1761-9, 2016 04 07.
Article En | MEDLINE | ID: mdl-26755710

A global phase 3 study evaluated the pharmacokinetics, efficacy, and safety of recombinant fusion protein linking coagulation factor IX with albumin (rIX-FP) in 63 previously treated male patients (12-61 years) with severe hemophilia B (factor IX [FIX] activity ≤2%). The study included 2 groups: group 1 patients received routine prophylaxis once every 7 days for 26 weeks, followed by either 7-, 10-, or 14-day prophylaxis regimen for a mean of 50, 38, or 51 weeks, respectively; group 2 patients received on-demand treatment of bleeding episodes for 26 weeks and then switched to a 7-day prophylaxis regimen for a mean of 45 weeks. The mean terminal half-life of rIX-FP was 102 hours, 4.3-fold longer than previous FIX treatment. Patients maintained a mean trough of 20 and 12 IU/dL FIX activity on prophylaxis with rIX-FP 40 IU/kg weekly and 75 IU/kg every 2 weeks, respectively. There was 100% reduction in median annualized spontaneous bleeding rate (AsBR) and 100% resolution of target joints when subjects switched from on-demand to prophylaxis treatment with rIX-FP (P< .0001). The median AsBR was 0.00 for all prophylaxis regimens. Overall, 98.6% of bleeding episodes were treated successfully, including 93.6% that were treated with a single injection. No patient developed an inhibitor, and no safety concerns were identified. These results indicate rIX-FP is safe and effective for preventing and treating bleeding episodes in patients with hemophilia B at dosing regimens of 40 IU/kg weekly and 75 IU/kg every 2 weeks. This trial was registered at www.clinicaltrials.gov as #NCT0101496274.


Albumins/administration & dosage , Albumins/pharmacokinetics , Factor IX/administration & dosage , Factor IX/pharmacokinetics , Hemophilia B/blood , Hemophilia B/prevention & control , Adolescent , Adult , Albumins/adverse effects , Child , Factor IX/adverse effects , Hemophilia B/pathology , Hemorrhage/blood , Hemorrhage/chemically induced , Humans , Male , Middle Aged , Prospective Studies , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/pharmacokinetics
12.
Thromb Haemost ; 115(1): 40-50, 2016 Jan.
Article En | MEDLINE | ID: mdl-26245874

The diagnosis of von Willebrand disease (VWD) remains difficult in a significant proportion of patients. A Spanish multicentre study investigated a cohort of 556 patients from 330 families who were analysed centrally. VWD was confirmed in 480. Next generation sequencing (NGS) of the whole coding VWF was carried out in all recruited patients, compared with the phenotype, and a final diagnosis established. A total of 238 different VWF mutations were found, 154 were not included in the Leiden Open Variation Database (LOVD). Of the patients, 463 were found to have VWF mutation/s. A good phenotypic/genotypic association was estimated in 96.5% of the patients. One hundred seventy-four patients had two or more mutations. Occasionally a predominant phenotype masked the presence of a second abnormality. One hundred sixteen patients presented with mutations that had previously been associated with increased von Willebrand factor (VWF) clearance. RIPA unavailability, central phenotypic results disagreement and difficult distinction between severe type 1 and type 3 VWD prevented a clear diagnosis in 70 patients. The NGS study facilitated an appropriate classification in 63 of them. The remaining seven patients presented with a VWF novel mutation pending further investigation. In five patients with a type 3 and two with a type 2A or 2B phenotype with no mutation, an acquired von Willebrand syndrome (AVWS) was suspected/confirmed. These data seem to support NGS as a first line efficient and faster paradigm in VWD diagnosis.


Mutation , von Willebrand Diseases/epidemiology , von Willebrand Diseases/genetics , von Willebrand Factor/genetics , Case-Control Studies , DNA Mutational Analysis/methods , Female , Genetic Association Studies , Genetic Markers , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Male , Molecular Epidemiology , Phenotype , Predictive Value of Tests , Registries , Risk Factors , Spain , von Willebrand Diseases/diagnosis
14.
Orphanet J Rare Dis ; 8: 170, 2013 Oct 20.
Article En | MEDLINE | ID: mdl-24139637

BACKGROUND: Mutations in PMM2 impair phosphomannomutase-2 activity and cause the most frequent congenital disorder of glycosylation, PMM2-CDG. Mannose-1-phosphate, that is deficient in this disorder, is also implicated in the biosynthesis of glycosylphosphatidyl inositol (GPI) anchors. OBJECTIVE: To evaluate whether GPI-anchor and GPI-anchored proteins are defective in PMM2-CDG patients. METHODS: The expression of GPI-anchor and seven GPI-anchored proteins was evaluated by flow cytometry in different cell types from twelve PMM2-CDG patients. Additionally, neutrophil CD16 and plasma hepatic proteins were studied by Western blot. Transferrin glycoforms were evaluated by HPLC. RESULTS: Patients and controls had similar surface expression of GPI-anchor and most GPI-anchored proteins. Nevertheless, patients displayed a significantly diminished binding of two anti-CD16 antibodies (3G8 and KD1) to neutrophils and also of anti-CD14 (61D3) to monocytes. Interestingly, CD16 immunostaining and asialotransferrin levels significantly correlated with patients' age. Analysis by flow cytometry of CD14 with MΦP9, and CD16 expression in neutrophils by Western blot using H-80 ruled out deficiencies of these antigens. CONCLUSIONS: PMM2 mutations do not impair GPI-anchor or GPI-anchored protein expression. However, the glycosylation anomalies caused by PMM2 mutations might affect the immunoreactivity of monoclonal antibodies and lead to incorrect conclusions about the expression of different proteins, including GPI-anchored proteins. Neutrophils and monocytes are sensitive to PMM2 mutations, leading to abnormal glycosylation in immune receptors, which might potentially affect their affinity to their ligands, and contribute to infection. This study also confirms less severe hypoglycosylation defects in older PMM2-CDG patients.


Glycosylphosphatidylinositols/metabolism , Phosphotransferases (Phosphomutases)/metabolism , Adolescent , Child , Child, Preschool , Chromatography, High Pressure Liquid , Congenital Disorders of Glycosylation , Female , Flow Cytometry , Glycosylphosphatidylinositols/genetics , Humans , Infant , Male , Monocytes/metabolism , Mutation , Neutrophils/metabolism , Phosphotransferases (Phosphomutases)/genetics
15.
Blood ; 121(20): 4046-55, 2013 May 16.
Article En | MEDLINE | ID: mdl-23553768

The objective of this study was to examine the association of the intensity of treatment, ranging from high-dose intensive factor VIII (FVIII) treatment to prophylactic treatment, with the inhibitor incidence among previously untreated patients with severe hemophilia A. This cohort study aimed to include consecutive patients with a FVIII activity < 0.01 IU/mL, born between 2000 and 2010, and observed during their first 75 FVIII exposure days. Intensive FVIII treatment of hemorrhages or surgery at the start of treatment was associated with an increased inhibitor risk (adjusted hazard ratio [aHR], 2.0; 95% confidence interval [CI], 1.3-3.0). High-dose FVIII treatment was associated with a higher inhibitor risk than low-dose FVIII treatment (aHR, 2.3; 95% CI, 1.0-4.8). Prophylaxis was only associated with a decreased overall inhibitor incidence after 20 exposure days of FVIII. The association with prophylaxis was more pronounced in patients with low-risk F8 genotypes than in patients with high-risk F8 genotypes (aHR, 0.61, 95% CI, 0.19-2.0 and aHR, 0.85, 95% CI, 0.51-1.4, respectively). In conclusion, our findings suggest that in previously untreated patients with severe hemophilia A, high-dosed intensive FVIII treatment increases inhibitor risk and prophylactic FVIII treatment decreases inhibitor risk, especially in patients with low-risk F8 mutations.


Blood Coagulation Factor Inhibitors/metabolism , Factor VIII/administration & dosage , Factor VIII/antagonists & inhibitors , Hemophilia A/drug therapy , Hemophilia A/epidemiology , Hemorrhage/prevention & control , Adolescent , Adult , Blood Coagulation Factor Inhibitors/blood , Chemoprevention/adverse effects , Child , Cohort Studies , Dose-Response Relationship, Drug , Hemophilia A/blood , Hemophilia A/metabolism , Hemorrhage/blood , Hemorrhage/epidemiology , Hemorrhage/metabolism , Humans , Risk Factors , Severity of Illness Index , Young Adult
16.
Haematologica ; 98(4): 538-44, 2013 Apr.
Article En | MEDLINE | ID: mdl-23403322

Because of the very short half-life of factor VII, prophylaxis in factor VII deficiency is considered a difficult endeavor. The clinical efficacy and safety of prophylactic regimens, and indications for their use, were evaluated in factor VII-deficient patients in the Seven Treatment Evaluation Registry. Prophylaxis data (38 courses) were analyzed from 34 patients with severe factor VII deficiency (<1-45 years of age, 21 female). Severest phenotypes (central nervous system, gastrointestinal, joint bleeding episodes) were highly prevalent. Twenty-one patients received recombinant activated factor VII (24 courses), four received plasma-derived factor VII, and ten received fresh frozen plasma. Prophylactic schedules clustered into "frequent" courses (three times weekly, n=23) and "infrequent" courses (≤ 2 times weekly, n=15). Excluding courses for menorrhagia, "frequent" and "infrequent" courses produced 18/23 (78%) and 5/12 (41%) "excellent" outcomes, respectively; relative risk, 1.88; 95% confidence interval, 0.93-3.79; P=0.079. Long term prophylaxis lasted from 1 to >10 years. No thrombosis or new inhibitors occurred. In conclusion, a subset of patients with factor VII deficiency needed prophylaxis because of severe bleeding. Recombinant activated factor VII schedules based on "frequent" administrations (three times weekly) and a 90 µg/kg total weekly dose were effective. These data provide a rationale for long-term, safe prophylaxis in factor VII deficiency.


Factor VII Deficiency/prevention & control , Factor VII/therapeutic use , Factor VIIa/therapeutic use , Plasma , Adolescent , Adult , Child , Child, Preschool , Factor VIIa/genetics , Female , Humans , Infant , Male , Middle Aged , Recombinant Proteins/therapeutic use , Registries/statistics & numerical data , Time Factors , Treatment Outcome , Young Adult
17.
AIDS Rev ; 14(4): 279-89, 2012.
Article En | MEDLINE | ID: mdl-23258302

During the last 25 years the safety of blood products has improved dramatically with regard to infectious risk, notably to the threat represented by retroviruses (HIV and human T­cell lymphotropic virus) and hepatitis B and C viruses. However, both residual and emergent viral infections are still responsible for contaminations in recipients of blood products. Along with other viruses (human herpesvirus­8, human parvovirus B19, hepatitis A and E viruses, etc.), special attention has recently been paid to emerging arboviruses, such as West Nile virus in North America, and Dengue and Chikungunya viruses in Europe. Another blood­linked risk, notably in the United Kingdom and France, is the prion agent responsible for the variant form of the Creutzfeldt­Jakob disease. Hemophilia care has been the model for improvements in the safety and availability of safe blood components free of infectious agents. In this regard, several measures aimed to halt transmission of viruses have been implemented in blood banks, including the exclusion of at­risk donors, specific sensitive diagnostic tests, leukocyte reduction of labile blood products, and the physical or chemical treatments aiming at nonspecific inactivation of infectious agents potentially present in blood without impairing significantly its physiological properties.


Blood Banks/standards , Blood Donors , Blood Safety , Communicable Diseases, Emerging/transmission , Communicable Diseases, Emerging/virology , Leukocyte Reduction Procedures , Virus Diseases/prevention & control , Virus Diseases/transmission , Arbovirus Infections/prevention & control , Arbovirus Infections/transmission , Blood Component Transfusion , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/prevention & control , Creutzfeldt-Jakob Syndrome/prevention & control , Creutzfeldt-Jakob Syndrome/transmission , Female , Humans , Male , Retroviridae Infections/prevention & control , Retroviridae Infections/transmission , Risk Factors
18.
Adv Ther ; 29 Suppl 1: 1-16, 2012 Apr.
Article En | MEDLINE | ID: mdl-22467446

The World Federation of Hemophilia (WFH) 2010 World Congress held in Buenos Aires, Argentina, in July 2010, attracted more than 4,300 participants from 106 countries. This report summarizes two symposia held during the congress. The first, titled "Emerging Co-Morbidities in the Aging Hemophilia Population: Healthcare Challenges and Treatment Opportunities," chaired by Gerry Dolan, MD, and Jussara Almeida Cruz, MD, examined the co-morbidities experienced by the aging hemophilic patient population, such as cardiovascular disease, cancer, arthritis, osteoporosis, hypertension, and obesity. In addition, Bayer's products in preclinical and clinical development were reviewed, including a novel factor VIIa variant and a long-acting factor VIII molecule, i.e., one that has undergone site-specific PEGylation (attachment of polyethylene glycol [PEG] polymer chains to another molecule). The other symposium, titled "Practical Steps to Making Better Care for Hemophilia Patients a Reality," chaired by Carmen Altisent, MD, and Cesar Guerrero, RN, reviewed the steps that hemophilia caregivers can take to improve the care of their patients. Issues such as the treatment of hemarthroses, the role of the research nurse, and the management of pediatric patients transitioning to adulthood were discussed.


Hemophilia A/therapy , Adolescent , Aged , Blood Coagulation Factors/chemistry , Blood Coagulation Factors/pharmacology , Blood Coagulation Factors/therapeutic use , Comorbidity , Drug Discovery , Geriatric Assessment , Global Health , Hemarthrosis/therapy , Hemophilia A/drug therapy , Hemophilia A/epidemiology , Humans , Life Expectancy , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Transition to Adult Care
19.
Haematologica ; 97(7): 1003-7, 2012 Jul.
Article En | MEDLINE | ID: mdl-22315491

Genetic analysis of von Willebrand disease by von Willebrand factor gene sequencing has not yet become routine practice. Nevertheless, the prospects for molecular diagnosis have changed dramatically in recent years with the unveiling of next-generation sequencing platforms. With the goal of applying this technology to von Willebrand disease, we designed a strategy for von Willebrand factor gene enrichment and multiplexing based on short polymerase chain reactions. Forty patients were simultaneously analyzed enabling the identification of 43 mutations, including 36 substitutions, 2 intronic splice site mutations, 2 indels, and 3 deletions. By pooling patient genomic DNA before polymerase chain reaction enrichment, indexing samples with barcode tags, and re-sequencing on the next-generation sequencing instrument, at least 350 patients and relatives per run can be simultaneously analyzed in a fast, inexpensive manner. This is one of the first reports in which this technology has been shown to be feasible for large-scale mutation screening by single gene re-sequencing.


Molecular Diagnostic Techniques/methods , Mutation , Sequence Analysis, DNA/methods , von Willebrand Diseases , von Willebrand Factor/genetics , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans , Multiplex Polymerase Chain Reaction , von Willebrand Diseases/diagnosis , von Willebrand Diseases/genetics
20.
Am J Pathol ; 178(6): 2938-48, 2011 Jun.
Article En | MEDLINE | ID: mdl-21641410

Clinical evidence accumulated from hemophilic patients during prophylaxis with recombinant activated factor VII (rFVIIa) suggests that the duration of the hemostatic action of rFVIIa exceeds its predicted plasma half-life. Mechanisms involved in this outcome have not been elucidated. We have investigated in vitro the redistribution of rFVIIa in platelets from healthy donors, patients with FVII deficiency, and one patient with Bernard-Soulier syndrome. Platelet-rich plasma was exposed to rFVIIa (3 to 60 µg/mL). Flow cytometry, immunocytochemistry, and coagulation tests were applied to detect and quantify rFVIIa. The hemostatic effect of rFVIIa associated to platelets was evaluated using perfusion models. Our studies revealed a dose-dependent association of rFVIIa to the platelet cytoplasm with redistribution into the open canalicular system, and α granules. Mechanisms implicated in the internalization are multiple, involve GPIb and GPIV, and require phospholipids and cytoskeletal assembly. After platelet activation with thrombin, platelets exposed rFVIIa on their membrane. Perfusion studies revealed that the presence of 30% of platelets containing FVIIa improved platelet aggregate formation and enhanced fibrin generation (P < 0.01 versus control). Our results indicate that, at therapeutic concentrations, rFVIIa can be internalized into platelets, where it is protected from physiological clearance mechanisms and can still promote hemostatic activity. Redistribution of rFVIIa into platelets may explain the prolonged prophylactic effectiveness of rFVIIa in hemophilia.


Blood Platelets/drug effects , Factor VIIa/metabolism , Hemostasis/drug effects , Hemostatics/pharmacology , Bernard-Soulier Syndrome/pathology , Bernard-Soulier Syndrome/physiopathology , Blood Platelets/ultrastructure , Case-Control Studies , Cell Extracts , Cell Membrane/drug effects , Cell Membrane/metabolism , Endocytosis/drug effects , Factor VIIa/pharmacology , Flow Cytometry , Humans , Platelet Activation/drug effects , Protein Transport/drug effects , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Thrombelastography , Thrombin/pharmacology , Tissue Donors
...