Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Haemophilia ; 30(1): 59-67, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37902395

ABSTRACT

INTRODUCTION: Hereditary factor X (FX) deficiency (HFXD) is an autosomal recessive rare bleeding disorder that leads to defects in the FX protein. Depending on the degree of deficiency, patients may be at risk of life-threatening bleeding episodes. Historical treatments for FX deficiency include prothrombin complex concentrates, which can increase the risk of thrombosis, and fresh frozen plasma, which can cause volume overload and transfusion reactions. Plasma-derived FX (pdFX), a single-factor, high-purity, high-potency human FX treatment, was approved in 2015 in the United States and in 2016 in Europe for on-demand treatment and prophylaxis of bleeding episodes and perioperative management of patients with HFXD. METHODS: Five studies that examined the use of pdFX in patients with mild (plasma FX activity [FX:C] ≥5 IU/dL), moderate (FX:C ≥1 and <5 IU/dL), or severe (FX:C < 1 IU/dL) HFXD were reviewed: TEN01, TEN02 and TEN03 were prospective, open-label, multicentre, nonrandomised studies, and TEN05 and TEN06 were multicentre retrospective studies. RESULTS: When used as an on-demand treatment, pdFX was judged by investigators to be successful in treating 41/42 (97.6%), 2/3 (66.6%) and 79/79 (100%) bleeds in TEN01, TEN02 and TEN05, respectively. When used prophylactically, pdFX was judged 'excellent' for the prevention of bleeds in nine (100%) and eight (100%) patients in TEN02 and TEN05, respectively. Perioperative treatment and pharmacokinetics were also assessed. pdFX was safe and well tolerated. CONCLUSIONS: Together, these studies support the use of pdFX for on-demand treatment of bleeding, routine prophylaxis, and perioperative management of bleeding in patients with HFXD.


Subject(s)
Factor X Deficiency , Factor X , Humans , Factor X/therapeutic use , Factor X/adverse effects , Factor X Deficiency/complications , Factor X Deficiency/drug therapy , Prospective Studies , Retrospective Studies , Hemorrhage/etiology , Hemorrhage/prevention & control , Plasma
2.
Haemophilia ; 29(6): 1394-1409, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37766495

ABSTRACT

INTRODUCTION: Hereditary factor X deficiency is a rare bleeding disorder, with limited treatment options. This paper describes the approach to pre-clinical development and characterization of a high-purity plasma-derived factor X concentrate, to achieve orphan drug marketing authorization for the treatment of hereditary factor X deficiency. METHODS: A chromatographic process was developed, to purify factor X from human plasma for fractionation. The product was characterized using in vitro, in vivo and ex vivo tests for potency, purity, thrombogenicity, immunogenicity, toxicity and stability. RESULTS: The production process complied with good pharmaceutical manufacturing practice. It achieved 6000-fold purification and 100-fold concentration of the factor X protein compared to human plasma. The factor X protein was 94%-96% pure. Other residual plasma proteins were well below levels in plasma, minimizing potential interference in hemostasis after therapeutic administration. Effective virus-reduction during manufacture, and the absence of thrombogenicity, toxicity and immunogenic potential were confirmed, addressing the main safety concerns historically associated with plasma-derived therapeutics. The freeze-dried product remained stable between +2°C and +30°C for at least three years. After reconstitution with water for injections, the factor X activity was maintained for at least 48 h at +18°C to +22°C. CONCLUSION: Targeted pre-clinical development of the first highly-purified concentrate to treat hereditary factor X deficiency is described. Following international guidelines for nonclinical safety testing, particular strategies were adopted for unmodified products derived from human blood plasma. This approach may also be relevant to the development of other ultra-orphan medicinal products.


Subject(s)
Factor X Deficiency , Factor X , Humans , Factor X/therapeutic use , Factor X Deficiency/drug therapy , Factor X Deficiency/complications , Hemorrhage/complications , Plasma , Pharmaceutical Preparations
3.
Intern Med ; 62(16): 2401-2406, 2023.
Article in English | MEDLINE | ID: mdl-37587057

ABSTRACT

Coagulation factor X (FX) deficiency causes severe hemorrhagic symptoms. We herein report a 90-year-old man with hemorrhagic symptoms and prolongation of prothrombin time (PT) and activated partial thromboplastin time (APTT). Cross-mixing tests showed a factor deficiency pattern, but administration of plasma products was not effective. Acquired coagulation factor deficiency was suspected, and immunosuppressive therapy was started. After the intervention, his hemorrhagic symptoms improved. A decrease in FX activity was later confirmed, and anti-FX autoantibody was retrospectively detected by an enzyme-linked immunosorbent assay. Immediate intervention is important for patients suspected of having acquired coagulation factor deficiency.


Subject(s)
Factor X Deficiency , Male , Humans , Aged, 80 and over , Factor X Deficiency/drug therapy , Retrospective Studies , Adrenal Cortex Hormones , Autoantibodies , Blood Coagulation Factors
4.
Haemophilia ; 28(4): 523-531, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35499465

ABSTRACT

INTRODUCTION: Hereditary factor X (FX) deficiency (FXD) is a rare autosomal recessive bleeding disorder. Plasma-derived FX (pdFX) is a high-purity FX concentrate approved in the United States and Europe for the treatment and prophylaxis of bleeding episodes and for peri-operative management in patients with hereditary FXD (HFXD). AIM: To review pharmacokinetic dosing, efficacy, and safety data for pdFX as routine prophylaxis for HFXD. METHODS: Summary of the published pharmacokinetic and safety data from TEN01, TEN02, TEN05, and real-world publications of pdFX for prophylaxis. RESULTS: Pharmacokinetic modelling data from the phase 3 TEN01 study supported administration of pdFX 25 IU/kg twice weekly for routine prophylaxis in adolescents/adults (aged ≥12 years). Results from nine paediatric patients in the phase 3 TEN02 study and eight adolescents/adults (aged ≥12 years) in the retrospective data-collection TEN05 study, along with real-world evidence, showed that routine prophylaxis with pdFX ≈40 IU/kg twice weekly in patients aged <12 years and pdFX ≈25 IU/kg twice weekly in patients aged ≥12 years was effective in bleeding prevention. CONCLUSIONS: pdFX was well tolerated in clinical studies, with no new safety signals identified during routine prophylactic use. Based on current evidence, it is recommended that routine prophylaxis with pdFX be initiated at 25 IU/kg twice weekly in adults/adolescents ≥12 years of age, and at a dosage of 40 IU/kg twice weekly in children <12 years of age. Thereafter, FX levels should be closely monitored, and dosages should be adjusted according to clinical response and to maintain trough levels ≥5 IU/dl.


Subject(s)
Factor X Deficiency , Factor X , Adolescent , Adult , Blood Coagulation Tests , Child , Clinical Trials, Phase III as Topic , Factor X/adverse effects , Factor X Deficiency/drug therapy , Hemorrhage/drug therapy , Hemorrhage/prevention & control , Humans , Retrospective Studies
5.
BMJ Case Rep ; 14(4)2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33827877

ABSTRACT

We report a case of progressive light-chain amyloidosis (otherwise known as AL amyloidosis) with acquired factor X (aFX) deficiency with a complete haematological response and rapid normalisation of FX levels following daratumumab monotherapy. To our knowledge, this is the first case report documenting successful treatment with daratumumab of aFX deficiency secondary to AL amyloidosis. The patient responded well to this therapy, with excellent symptomatic and quality of life improvements as well as a reduction in bleeding manifestations. This case highlights the value in considering daratumumab treatment when AL amyloidosis is complicated by FX deficiency.


Subject(s)
Amyloidosis , Factor X Deficiency , Immunoglobulin Light-chain Amyloidosis , Amyloidosis/drug therapy , Factor X Deficiency/complications , Factor X Deficiency/drug therapy , Hemorrhage , Humans , Immunoglobulin Light-chain Amyloidosis/drug therapy , Quality of Life
6.
A A Pract ; 15(2): e01405, 2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33577168

ABSTRACT

Factor X (FX) deficiency is a rare coagulopathy that may cause bleeding complications in parturients. The literature on rotational thromboelastometry (ROTEM; Instrumentation Laboratory, Bedford, MA) to guide factor repletion and neuraxial placement during partuition is limited. We present a 17-year-old parturient with FX deficiency after vaginal delivery with neuraxial anesthesia without bleeding complications. After FX concentrate administration, thromboelastometry was utilized to risk-stratify and manage her coagulopathy peripartum. Thromboelastometry may be a valuable adjunct to conventional monitoring in patient management. A limitation of this report is that coagulation tests and thromboelastometry were not assessed at identical timepoints.


Subject(s)
Blood Coagulation Disorders , Factor X Deficiency , Adolescent , Blood Coagulation Tests , Delivery, Obstetric , Factor X Deficiency/drug therapy , Female , Humans , Pregnancy , Thrombelastography
7.
J Thromb Haemost ; 18(10): 2551-2556, 2020 10.
Article in English | MEDLINE | ID: mdl-32613702

ABSTRACT

BACKGROUND: Congenital factor X deficiency (FXD) is a rare bleeding disorder that often presents with severe bleeding in the neonatal period. Long-term prophylaxis with infusions of FX-containing products is recommended in patients with FXD and a personal or family history of severe bleeding. A plasma-derived FX concentrate (pdFX) is approved for on-demand and prophylactic therapy in adults and children with FXD. The safety and efficacy of pdFX has been demonstrated in patients <12 years of age, yet limited data exist regarding its use in infants. PATIENTS/METHODS: This retrospective case series details clinical experience using pdFX in four neonates with moderate and severe FXD across four institutions. RESULTS AND CONCLUSIONS: All four patients presented in the first week of life with severe bleeding. Following treatment of the acute bleed, prophylactic pdFX was initiated at an average of 29 days of life and a dose of 69 IU/kg every 48 hours. Incremental recovery (IR) in three infants averaged 1.42 IU/dL per IU/kg (min-max: 1.06-1.67 IU/dL per IU/kg). One patient experienced thrombotic complications in the setting of sepsis. After a median follow-up of 26.5 months, no patient has experienced breakthrough bleeding episodes. Our study supports the use of pdFX in neonates and infants and suggests that higher pdFX dosing of 70 to 80 IU/kg every 48 hours based on the smallest available vial size is feasible. Because of variability in IR, close monitoring of FX activity should be used to guide dosing in this age group.


Subject(s)
Factor X Deficiency , Factor X , Adult , Blood Coagulation Tests , Child , Factor X Deficiency/diagnosis , Factor X Deficiency/drug therapy , Female , Hemorrhage/chemically induced , Humans , Infant , Infant, Newborn , Retrospective Studies
8.
Int J Hematol ; 111(1): 51-56, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31667683

ABSTRACT

Congenital factor X (FX) deficiency is a rare bleeding disorder with an incidence of one in one million. The proband, a 2-year-old girl, exhibited easy bruising and a history of umbilical cord bleeding at birth. Prothrombin time (> 40 s) and activated partial thromboplastin time (65.0 s) were prolonged. Marked declines in FX activity (< 1%) and FX antigen levels (5%) were also observed. Genetic analysis of the proband identified two types of single-base substitutions, c.353G>A (p.Gly118Asp) and c.1303G>A (p.Gly435Ser), indicating compound heterozygous congenital FX deficiency. Genetic analysis of family members revealed that her father and older sister (5-year-old) were also heterozygous for p.Gly118Asp, and that her mother was heterozygous for p.Gly435Ser. To improve the bleeding tendency, the proband received regular replacement of 500 units of PPSB-HT, a prothrombin complex concentrate (PCC). Following continued regular replacement of 500 units of PPSB-HT once per week, the proband has exhibited no bleeding tendencies and no new bruises have been observed. There are no previous report of the use of PPSB-HT for regular FX replacement. Regular replacement therapy with PPSB-HT may be an effective method for preventative control of bleeding tendencies in FX deficiency.


Subject(s)
Blood Coagulation Factors/therapeutic use , Factor X Deficiency/drug therapy , Factor X Deficiency/genetics , Factor X/genetics , Adult , Child, Preschool , Factor X/metabolism , Factor X Deficiency/enzymology , Factor X Deficiency/pathology , Female , Genetic Testing , Genotype , Hemorrhage/genetics , Heterozygote , Humans , Male , Mutation, Missense , Partial Thromboplastin Time , Pedigree , Phenotype , Prothrombin Time
10.
J Investig Med High Impact Case Rep ; 7: 2324709619832332, 2019.
Article in English | MEDLINE | ID: mdl-30947547

ABSTRACT

Acquired factor X (FX) deficiency is a rare but serious complication of primary amyloidosis, presumably caused by the binding of amyloid proteins to the clotting factors. The prolonged prothrombin time, partial thromboplastin time, and low FX level, which are correctable by mixing study, are the disease hallmarks. An immediate goal of care is to stop bleeding. Clotting factor replacement requires close monitoring of coagulogram and FX levels due to varying FX clearance among patients. High-purity FX is currently approved for hereditary FX deficiency and has been successfully used in some acquired FX deficiency cases. Ongoing bleeding risk complicates the treatment decision. Novel therapies yielding rapid and deep response reduce amyloid protein production and improve long-term outcome.


Subject(s)
Blood Coagulation Factors/administration & dosage , Factor X Deficiency/complications , Factor X/administration & dosage , Hemorrhage/etiology , Immunoglobulin Light-chain Amyloidosis/complications , Adult , Aged , Amyloidogenic Proteins/metabolism , Antineoplastic Agents/therapeutic use , Factor X Deficiency/blood , Factor X Deficiency/drug therapy , Female , Hemorrhage/drug therapy , Humans , Immunoglobulin Light-chain Amyloidosis/drug therapy , Immunosuppressive Agents/therapeutic use , Male , Partial Thromboplastin Time
11.
Melanoma Res ; 29(1): 99-101, 2019 02.
Article in English | MEDLINE | ID: mdl-30376464

ABSTRACT

Malignant melanoma is very rare in childhood. The approach to diagnosis and treatment in children has been adopted from adult guidelines. Vemurafenib is indicated in adults with BRAF V600 mutation-positive stage IIIc/IV melanoma and causes cutaneous adverse events. We report on a 3-year-old child with recurrent, metastatic (bone) BRAF mutation-positive melanoma. He also had severe factor X deficiency. Four days after vemurafenib treatment, bilateral palpebral edema and violet-colored hyperpigmentation were observed. There was no objective response to vemurafenib; however, bone pain regressed slightly. Our patient is the youngest patient who received vemurafenib for BRAF V600 mutation-positive metastatic melanoma in the literature.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/etiology , Factor X Deficiency/drug therapy , Melanoma/drug therapy , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/drug therapy , Vemurafenib/adverse effects , Child, Preschool , Disease Progression , Drug-Related Side Effects and Adverse Reactions/pathology , Factor X Deficiency/complications , Factor X Deficiency/genetics , Factor X Deficiency/pathology , Humans , Male , Melanoma/complications , Melanoma/genetics , Melanoma/pathology , Mutation , Prognosis , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/complications , Skin Neoplasms/genetics , Skin Neoplasms/secondary
12.
J Thromb Haemost ; 16(5): 849-857, 2018 05.
Article in English | MEDLINE | ID: mdl-29460388

ABSTRACT

Essentials Plasma-derived factor X concentrate (pdFX) is used to treat hereditary factor X deficiency. pdFX pharmacokinetics, safety and efficacy were assessed in factor X-deficient women/girls. Treatment success rate was 98%; only 6 adverse events in 2 subjects were possibly pdFX related. On-demand pdFX 25 IU kg-1 was effective and safe in women/girls with factor X deficiency. SUMMARY: Background A high-purity, plasma-derived factor X concentrate (pdFX) has been approved for the treatment of hereditary FX deficiency, an autosomal recessive disorder. Objective To perform post hoc assessments of pdFX pharmacokinetics, safety and efficacy in women and girls with hereditary FX deficiency. Patients/Methods Subjects aged ≥ 12 years with moderate/severe FX deficiency (plasma FX activity of < 5 IU dL-1 ) received on-demand or preventive pdFX (25 IU kg-1 ) for ≤ 2 years. Results Of 16 enrolled subjects, 10 women and girls (aged 14-58 years [median, 25.5 years]) received 267 pdFX infusions. Mean monthly infusions per subject were higher among women and girls (2.48) than among men and boys (1.62). In women and girls, 132 assessable bleeding episodes (61 heavy menstrual bleeds, 47 joint bleeds, 15 muscle bleeds, and nine other bleeds) were treated with pdFX, with a 98% treatment success rate versus 100% in men and boys. Mean pdFX incremental recovery was similar in the two groups (2.05 IU dL-1 versus 1.91 IU dL-1 per IU kg-1 ), as was the mean half-life (29.3 h versus 29.5 h). Of 142 adverse events in women and girls, headache was the most common (12 events in six subjects). Six events (two infusion-site erythema, two fatigue, one back pain, one infusion-site pain) in two subjects were considered to be possibly pdFX-related. Following the trial, pdFX was used to successfully maintain hemostasis in two subjects undergoing obstetric delivery. Conclusions pdFX was well tolerated and effective in women and girls with FX deficiency. Although women and girls had different bleeding symptoms and sites than men and boys, their pdFX pharmacokinetic profile was comparable.


Subject(s)
Blood Coagulation/drug effects , Coagulants/pharmacokinetics , Factor X Deficiency/drug therapy , Factor X/pharmacokinetics , Hemorrhage/drug therapy , Adolescent , Adult , Age Factors , Blood Coagulation/genetics , Child , Coagulants/administration & dosage , Coagulants/adverse effects , Europe , Factor X/administration & dosage , Factor X/adverse effects , Factor X/genetics , Factor X Deficiency/blood , Factor X Deficiency/diagnosis , Factor X Deficiency/genetics , Genetic Predisposition to Disease , Hemorrhage/blood , Hemorrhage/diagnosis , Hemorrhage/genetics , Humans , Middle Aged , Patient Safety , Phenotype , Risk Assessment , Risk Factors , Severity of Illness Index , Sex Factors , Treatment Outcome , United States , Young Adult
13.
Expert Opin Drug Metab Toxicol ; 13(1): 97-104, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27797267

ABSTRACT

INTRODUCTION: Hereditary factor X (FX) deficiency is a rare autosomal recessive bleeding disorder characterized mainly by mild-to-severe bleeding into the mucous membranes, muscles or joints. Previously, treatment options for hereditary FX deficiency were limited mostly to products that may not specify FX content (i.e. fresh frozen plasma and prothrombin complex concentrates) and that have associated safety concerns. To meet the need for a single-factor replacement therapy specifically for use in FX-deficient patients, a high-purity, high-potency, human plasma-derived FX concentrate (pdFX; Coagadex®; Bio Products Laboratory, Elstree, UK) has been developed and approved for treatment of perioperative bleeding and on-demand treatment in FX-deficient patients. Areas covered: The pharmacology, efficacy, and safety of pdFX are discussed, with a review of preclinical studies and clinical trial data that led to regulatory approval of pdFX in the United States and Europe. Expert opinion: As the first single-factor replacement therapy indicated for hereditary FX deficiency, pdFX is a safe and efficacious treatment option in patients aged ≥12 years with hereditary FX deficiency. Clinical studies of pdFX provide a dosing regimen for use in cases of bleeding episodes, surgery, and prophylaxis. Further studies are ongoing regarding use of pdFX long term and in patients aged ≤12 years.


Subject(s)
Coagulants/administration & dosage , Factor X Deficiency/drug therapy , Factor X/administration & dosage , Animals , Blood Loss, Surgical/prevention & control , Coagulants/adverse effects , Coagulants/pharmacokinetics , Drug Evaluation, Preclinical/methods , Factor X/adverse effects , Factor X/pharmacokinetics , Factor X Deficiency/complications , Factor X Deficiency/physiopathology , Humans , Perioperative Care/methods
14.
Lab Med ; 47(3): e35-7, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27378481

ABSTRACT

Congenital factor X deficiency is an extremely rare coagulation disorder that can place patients at risk for spontaneous hemorrhage or excessive bleeding in the setting of trauma or invasive procedures. Given the rarity of this disorder, there is little published guidance on how best to prevent or treat bleeding. Herein, we report a case of a 56-year-old white man with congenital factor X deficiency who was scheduled for major neurosurgery and who was treated perioperatively with 4-factor prothrombin complex concentrate (4F-PCC). Doses of 4F-PCC at 15 U per kg, administered immediately preoperatively and once at 24 hours postoperatively, allowed for successful completion of an anterior cervical discectomy and fusion without excessive bleeding. Moreover, no thromboembolic complications were observed. As such, given the wide availability of 4F-PCC, it may be considered as a first-line therapy and an alternative to fresh frozen plasma for factor X deficiencies, particularly in high-risk operative cases.


Subject(s)
Blood Coagulation Factors/administration & dosage , Factor X Deficiency/drug therapy , Neurosurgery , Postoperative Care/methods , Preoperative Care/methods , Diskectomy , Humans , Male , Middle Aged , Spinal Fusion , Treatment Outcome
15.
Haemophilia ; 22(5): 713-20, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27217097

ABSTRACT

INTRODUCTION: Maintaining haemostasis in surgery is challenging for hereditary rare bleeding disorders in which multi-coagulation-factor concentrates are the only therapeutic option. Hereditary factor X (FX) deficiency affects 1:500 000 to 1:1 000 000 individuals, and no specific replacement FX concentrate has been available. A high-purity, plasma-derived FX concentrate (pdFX) has been developed for patients with hereditary FX deficiency. AIM: Our objective was to assess the safety and efficacy of pdFX in subjects with FX deficiency undergoing surgery. METHODS: Subjects with hereditary mild-to-severe FX deficiency (basal plasma FX activity [FX:C] <20 IU dL(-1) ) undergoing surgery received pdFX preoperatively to raise FX:C to 70-90 IU dL(-1) and postoperatively to maintain levels >50 IU dL(-1) until the subject was no longer at risk of bleeding due to surgery. Efficacy of pdFX was assessed by blood loss during surgery, requirement for blood transfusion, postoperative bleeding from the surgical or other sites, and changes in haemoglobin levels. Safety was assessed by adverse events (AEs), development of inhibitors, and clinically significant changes in laboratory parameters. RESULTS: Five subjects (aged 14-59 years) underwent seven surgical procedures (four major and three minor). Treatment duration was 1-15 days. For each procedure, pdFX treatment was assessed as "excellent" in preventing bleeding and achieving haemostasis. No blood transfusions were required, no AEs related to pdFX were observed, and no clinically significant trends were found in any laboratory parameters. CONCLUSION: These data demonstrate that pdFX is safe and effective as replacement therapy in five subjects with mild-to-severe FX deficiency undergoing surgery on seven occasions.


Subject(s)
Coagulants/therapeutic use , Factor X Deficiency/drug therapy , Factor X/therapeutic use , Adolescent , Adult , Coagulants/analysis , Coagulants/isolation & purification , Factor X/analysis , Factor X/isolation & purification , Factor X Deficiency/pathology , Female , Hemoglobins/analysis , Hemorrhage/prevention & control , Humans , Male , Middle Aged , Preoperative Care , Severity of Illness Index , Treatment Outcome , Young Adult
16.
Haemophilia ; 22(3): 419-25, 2016 May.
Article in English | MEDLINE | ID: mdl-27197801

ABSTRACT

INTRODUCTION: Hereditary factor X (FX) deficiency is a rare bleeding disorder affecting 1:500 000 to 1:1 000 000 of individuals. Until recently, no specific replacement factor concentrate was available. AIM: The aim of this study was to assess safety and efficacy of a new, high-purity plasma-derived FX concentrate (pdFX) in subjects with hereditary FX deficiency. METHODS: Subjects aged ≥12 years with moderate or severe FX deficiency (plasma FX activity <5 IU dL(-1) ) received 25 IU kg(-1) pdFX as on-demand treatment or short-term prophylaxis for 6 months to 2 years. Subjects assessed pdFX efficacy for each bleed; at end-of-study, investigators assessed overall pdFX efficacy. Blood samples for pharmacokinetic analysis were obtained at baseline and ≥6 months. Safety was assessed by adverse events (AEs), inhibitor development and changes in laboratory parameters. RESULTS: Sixteen enrolled subjects (six aged 12-17 years; 10 aged 18-58 years) received a total of 468 pdFX infusions. In the 187 analysed bleeds, pdFX efficacy was categorized as excellent, good, poor or unassessable in 90.9%, 7.5%, 1.1% and 0.5% of bleeds respectively; 83% of bleeds were treated with one infusion. For pdFX, mean (median; interquartile range) incremental recovery and half-life were 2.00 (2.12; 1.79-2.37) IU dL(-1) per IU kg(-1) and 29.4 (28.6; 25.8-33.1) h respectively. No serious AEs possibly related to pdFX or evidence of FX inhibitors were observed, and no hypersensitivity reactions or clinically significant trends were detected in laboratory parameters. CONCLUSION: These results demonstrate that a dose of 25 IU kg(-1) pdFX is safe and efficacious for on-demand treatment and short-term prophylaxis in subjects with moderate or severe hereditary FX deficiency.


Subject(s)
Factor X Deficiency/drug therapy , Factor X/therapeutic use , Adolescent , Adult , Antibodies, Neutralizing/blood , Blood Coagulation Tests , Child , Factor X/adverse effects , Factor X/pharmacokinetics , Factor X Deficiency/congenital , Factor X Deficiency/pathology , Female , Half-Life , Hemorrhage/prevention & control , Humans , Male , Menorrhagia/prevention & control , Middle Aged , Severity of Illness Index , Treatment Outcome , Young Adult
17.
Pediatr Blood Cancer ; 63(7): 1300-4, 2016 07.
Article in English | MEDLINE | ID: mdl-27098186

ABSTRACT

Factor X deficiency (FXD) is a rare bleeding disorder, which can result in severe bleeding symptoms such as intracranial hemorrhage (ICH). The most common bleeding symptoms are epistaxis and gum bleeding. ICH is reported in 9-26% of all patients with FXD, mostly during the first month of life. Here, we present a rare case of a male presenting with ICH at the age of 20 months as the first manifestation of FXD. Secondary prophylaxis with factor X substitution once weekly prevented further bleeding.


Subject(s)
Factor X Deficiency , Factor X/administration & dosage , Intracranial Hemorrhages , Factor X Deficiency/complications , Factor X Deficiency/diagnosis , Factor X Deficiency/drug therapy , Factor X Deficiency/pathology , Humans , Infant , Intracranial Hemorrhages/diagnosis , Intracranial Hemorrhages/etiology , Intracranial Hemorrhages/pathology , Intracranial Hemorrhages/prevention & control , Male
18.
Haemophilia ; 22(3): 426-32, 2016 May.
Article in English | MEDLINE | ID: mdl-26879266

ABSTRACT

INTRODUCTION: Hereditary factor X (FX) deficiency affects 1:500 000 to 1:1 000 000 of individuals. There are few published data on the pharmacokinetics (PK) of FX for existing treatments for FX deficiency, and no specific replacement factor concentrate exists. A high-purity plasma-derived FX concentrate (pdFX) has been developed for use as replacement therapy in subjects with hereditary FX deficiency. AIM: This analysis assessed pdFX PK after a single 25 IU kg(-1) bolus dose in subjects with hereditary moderate or severe FX deficiency (plasma FX activity [FX:C] <5 IU dL(-1) ). METHODS: For a baseline PK assessment, blood samples were taken predose and at intervals up to 144 h (7 days) post dose. After ≥6 months of on-demand pdFX treatment and treatment of ≥1 bleed with pdFX, subjects underwent repeat PK assessment. Samples were assayed for plasma FX:C (measured using the clotting and chromogenic assays) and FX antigen. RESULTS: FX:C peaked at 0.4-0.5 h and subsequently declined over the course of 144 h with a biphasic decay curve. PK parameters observed at the baseline (n = 16) and repeat (n = 15) assessments were equivalent, therefore summary PK values were obtained by combining data from both visits (n = 31). The mean terminal half-life and incremental recovery of pdFX was 29.4 h and 2.00 IU dL(-1) per IU kg(-1) respectively. CONCLUSION: This is the most comprehensive PK study to date in subjects with hereditary FX deficiency. These results are consistent with the observed haemostatic efficacy of pdFX and provide the PK data required for the treatment of hereditary FX deficiency using pdFX replacement therapy.


Subject(s)
Coagulants/therapeutic use , Factor X Deficiency/drug therapy , Factor X/therapeutic use , Adolescent , Adult , Area Under Curve , Blood Coagulation Tests , Child , Coagulants/pharmacokinetics , Factor X/pharmacokinetics , Factor X Deficiency/congenital , Factor X Deficiency/pathology , Female , Half-Life , Humans , Male , Middle Aged , Prospective Studies , ROC Curve , Severity of Illness Index , Treatment Outcome , Young Adult
19.
Eur J Haematol ; 96(1): 60-4, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25782416

ABSTRACT

Up to 14% of individuals with systemic AL amyloidosis develop acquired factor X deficiency, which occurs due to adsorption of factor X onto amyloid fibrils. Although baseline factor X levels are not predictive of bleeding risk in these patients, serious hemorrhagic complications can occur, particularly during invasive procedures. Optimal management strategies to attenuate bleeding risk in these patients are unknown. We describe our experience in the management of acquired factor X deficiency, secondary to systemic AL amyloidosis, in a case series of three patients who received prothrombin complex concentrates (PCCs) for treatment and prevention of bleeding events. We performed a retrospective review extracting information on baseline demographics, laboratory data, pharmacokinetic (PK) studies, and clinically documented bleeding events. Our case series demonstrates that individuals with acquired factor X deficiency secondary to amyloidosis have variable laboratory and clinical responses to PCCs. This is likely due to distinct amyloid loads and fibril sequences, leading to different binding avidities for factor X. Our data emphasize the importance of performing PK testing prior to any invasive procedures to determine the dose and frequency interval to achieve adequate factor X levels for hemostasis, given the variable response between individuals.


Subject(s)
Amyloidosis , Blood Coagulation Factors , Factor X Deficiency , Adult , Aged , Amyloidosis/blood , Amyloidosis/complications , Amyloidosis/drug therapy , Blood Coagulation Factors/administration & dosage , Blood Coagulation Factors/pharmacokinetics , Factor X Deficiency/blood , Factor X Deficiency/drug therapy , Factor X Deficiency/etiology , Female , Humans , Male , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...