1
|
de Kovel M, van Haaster AC, Carcao M, Ranta S, Glosli H, Rivard GE, Kenet G, Kurnik K, Van Geet C, Carvalho M, Andersson NG, Kartal-Kaess M, Ljung R, van den Berg HM. Blood Group O Does Not Increase the Risk of Inhibitors in Severe Haemophilia A: Data from the PedNet Study Group. Haemophilia 2025. [PMID: 40123267 DOI: 10.1111/hae.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/13/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Inhibitor development against factor VIII (FVIII) is the most common complication of hemophilia A replacement therapy. One of the variables considered to influence inhibitor development is the ABO blood group. Patients with blood group O have, on average, a 30%-40% lower endogenous von Willebrand factor (VWF) concentration. It has been postulated that VWF levels influence inhibitor development. The objective of this study was to investigate the inhibitor risk in patients with severe hemophilia A comparing those with blood group O with those with non-O blood groups. METHODS The study population consisted of children with severe hemophilia A, born between 2000 and 2020, who reached 50 FVIII exposure days in the PedNet registry. Inhibitors were considered to be clinically relevant when at least two consecutive measurements were tested positive. RESULTS Routine testing of blood groups varied between centres: Out of 1172 patients with severe hemophilia A, blood group status was known in 759 patients (65.8%). The relative risk of inhibitor development for blood group O in comparison to non-O was 1.04 (95% CI: 0.7-1.7). CONCLUSION In the PedNet cohort, blood group O did not increase the risk of inhibitors in previously untreated children with severe hemophilia A. TRIAL REGISTRATION PedNet Registry; clinicaltrials.gov identifier: NCT02979119.
Collapse
Affiliation(s)
| | | | - Manuel Carcao
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Susanna Ranta
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Heidi Glosli
- Centre for Rare Disorders Oslo University Hospital, Oslo, Norway
| | - Georges E Rivard
- Division of Hematology/Oncology, Hôpital St Justine, Montréal, Canada
| | - Gili Kenet
- National Hemophilia Center, Sheba Medical Center, Tel Hashomer and Amalia Biron Research Institute of Thrombosis and Hemostasis, Tel Aviv University, Tel Aviv, Israel
| | - Karin Kurnik
- Paediatric Thrombosis and Haemostasis Unit, Paediatric Haemophilia Center, Dr. von Hauner Children's Hospital, LMU Munich, Munich, Germany
| | - Christel Van Geet
- Department of Pediatrics, Pediatric Hematology and Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Manuela Carvalho
- Centro de Referência de Coagulopatias Congénitas do Centro Hospitalar Universitário S. João, EPE, Porto, Portugal
| | - Nadine G Andersson
- Center for Thrombosis and Haemostasis, Skåne University Hospital, Malmö, Sweden
- Department of Paediatric Haematology and Oncology, Skåne University Hospital, Lund, Sweden
- Department of Clinical Sciences and Paediatrics, Lund University, Lund, Sweden
| | - Mutlu Kartal-Kaess
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Rolf Ljung
- Department of Paediatric Haematology and Oncology, Skåne University Hospital, Lund, Sweden
| | | |
Collapse
|
2
|
Park YS, Yoo KY, Park SK, Hwang T, Jung A, Choi EJ. Evaluation of FVIII pharmacokinetic profiles in Korean hemophilia A patients assessed with myPKFiT: a retrospective chart review. Blood Res 2024; 59:29. [PMID: 39196490 PMCID: PMC11358568 DOI: 10.1007/s44313-024-00023-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/21/2024] [Indexed: 08/29/2024] Open
Abstract
PURPOSE This study aimed to investigate the pharmacokinetics (PK) of factor VIII (FVIII) in Korean patients, as limited information is available on the PK of FVIII in this population. METHODS We collected the FVIII PK results from patients with moderate-to-severe hemophilia A using myPKFiT. PK variations were assessed according to age, blood type, inhibitor history, von Willebrand factor antigen (vWF:Ag) level, and body mass index. Additionally, the correlation between the PK profile and prophylaxis regimen was specifically analyzed for each product in severe cases. RESULTS The PK data of 48 and 81 patients treated with octocog alfa and rurioctocog alfa pegol, respectively, were obtained. The median half-lives of octocog alfa and rurioctocog alfa pegol were 9.9 (range: 6.3-15.2) h and 15.3 (range: 10.4-23.9) h, respectively. The PK profiles for each product did not differ according to age group; however, blood type-O patients had shorter half-lives and time to 1% compared to non-blood type-O patients. In regression analysis, the PK of octocog alfa showed a statistically significant difference according to age, whereas the PK of rurioctocog alfa pegol correlated with vWF:Ag. Only the frequency of rurioctocog alfa pegol use showed a statistically significant difference in relation to time to 1%, although the coefficient of determination was small. CONCLUSION This study confirmed significant interpatient variation in the PK of FVIII among Korean patients with hemophilia A. To achieve optimized prophylaxis, personalizing the regimen based on the PK profile of each individual patient is essential.
Collapse
Affiliation(s)
- Young-Shil Park
- Department of Pediatrics, Kyung Hee University Hospital, Gangdong, Seoul, Republic of Korea
| | - Ki-Young Yoo
- Korea Hemophilia Foundation Clinic, Seoul, Republic of Korea
| | - Sang Kyu Park
- Korea Hemophilia Foundation Clinic, Busan, Republic of Korea
| | - Taiju Hwang
- Korea Hemophilia Foundation Clinic, Gwangju, Republic of Korea
| | - Aeran Jung
- Medical Affairs, Takeda Pharmaceuticals Korea Co., Ltd, Seoul, Republic of Korea
| | - Eun Jin Choi
- Department of Pediatrics, Daegu Catholic University Medical Center, 33 Duryugongwon-Ro 17-Gil, Nam-Gu, Daegu, 42472, Republic of Korea.
| |
Collapse
|
3
|
Horling FM, Reipert BM, Allacher P, Engl W, Pan L, Tangada S. Immunogenicity profile of rurioctocog alfa pegol in previously treated patients with severe congenital hemophilia A. Blood Adv 2024; 8:2726-2739. [PMID: 38564770 PMCID: PMC11170177 DOI: 10.1182/bloodadvances.2023011780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/22/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024] Open
Abstract
ABSTRACT Rurioctocog alfa pegol is an extended-half-life full-length recombinant factor VIII (FVIII) bound to 20-kDa polyethylene glycol (PEG) that has been shown to be well tolerated and efficacious in the treatment and prevention of bleeding events in previously treated patients with severe hemophilia A. Here, we present a comprehensive analysis of immunogenicity data collected during 6 clinical studies of rurioctocog alfa pegol, including a total of 360 unique previously treated patients with severe hemophilia A. The analysis included treatment-emerging FVIII-neutralizing antibodies (FVIII inhibitors); preexisting and treatment-emerging antibodies binding to FVIII, PEG-FVIII, or PEG; and treatment-emerging antibodies binding to Chinese hamster ovary host cell proteins. Moreover, the potential association between the presence of these binding antibodies and adverse events (AEs) observed in patients was investigated, and the potential impact of these antibodies on the incremental recovery of rurioctocog alfa pegol in patients was analyzed. Overall, the data indicate that rurioctocog alfa pegol is not associated with any unexpected immunogenicity characteristics. Of 360 patients, 1 patient developed a transient FVIII inhibitor with a titer of 0.6 Bethesda units per mL, which was not associated with any serious AEs. Antibodies binding to FVIII, PEG-FVIII, or PEG were not detected at the time when the inhibitor was present. Moreover, 54 of 360 patients either entered the clinical studies with preexisting binding antibodies or developed these antibodies after exposure to rurioctocog alfa pegol. These antibodies were transient in most patients and did not show any causal relationship to either AEs or spontaneous bleeding episodes.
Collapse
Affiliation(s)
- Frank M. Horling
- Institute Krems Bioanalytics, IMC University of Applied Sciences Krems, Krems, Austria
| | - Birgit M. Reipert
- Institute Krems Bioanalytics, IMC University of Applied Sciences Krems, Krems, Austria
| | - Peter Allacher
- Institute Krems Bioanalytics, IMC University of Applied Sciences Krems, Krems, Austria
| | - Werner Engl
- Baxalta Innovations GmbH, a Takeda company, Vienna, Austria
| | - Luying Pan
- Takeda Development Center Americas, Inc, Cambridge, MA
| | | |
Collapse
|
4
|
Westwood LJ, Le Couteur DG, Hunt NJ, Cogger VC. Strategies to target and genetically modify the liver sinusoid. SINUSOIDAL CELLS IN LIVER DISEASES 2024:161-189. [DOI: 10.1016/b978-0-323-95262-0.00008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
5
|
Swystun LL, Michels A, Lillicrap D. The contribution of the sinusoidal endothelial cell receptors CLEC4M, stabilin-2, and SCARA5 to VWF-FVIII clearance in thrombosis and hemostasis. J Thromb Haemost 2023; 21:2007-2019. [PMID: 37085036 PMCID: PMC11539076 DOI: 10.1016/j.jtha.2023.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/23/2023]
Abstract
Quantitative abnormalities in factor VIII (FVIII) and its binding partner, von Willebrand factor (VWF), are associated with an increased risk of bleeding or thrombosis, and pathways that regulate the clearance of VWF-FVIII can strongly influence their plasma levels. In 2010, the Cohorts for Heart and Aging Research in Genome Epidemiology (CHARGE) on genome-wide association study meta-analysis identified variants in the genes for the sinusoidal endothelial receptors C-type lectin domain family 4 member M (CLEC4M), stabilin-2, and scavenger receptor class A member 5 (SCARA5) as being associated with plasma levels of VWF and/or FVIII in normal individuals. The ability of these receptors to bind, internalize, and clear the VWF-FVIII complex from the circulation has now been reported in a series of studies using in vitro and in vivo models. The receptor stabilin-2 has also been shown to modulate the immune response to infused VWF-FVIII concentrates in a murine model. In addition, the influence of genetic variants in CLEC4M, STAB2, and SCARA5 on type 1 von Willebrand disease/low VWF phenotype, FVIII pharmacokinetics, and the risk of venous thromboembolism has been described in a number of patient-based studies. Understanding the role of these receptors in the regulation of VWF-FVIII clearance has led to significant insights into the genomic architecture that modulates plasma VWF and FVIII levels, improving the understanding of pathways that regulate VWF-FVIII clearance and the mechanistic basis of quantitative VWF-FVIII pathologies.
Collapse
Affiliation(s)
- Laura L Swystun
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Alison Michels
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada. https://twitter.com/michels_alison
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
6
|
Konkle BA. Efanesoctocog alfa for the prevention and treatment of bleeding in patients with hemophilia A. Expert Rev Hematol 2023; 16:567-573. [PMID: 37289594 DOI: 10.1080/17474086.2023.2223925] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/06/2023] [Indexed: 06/10/2023]
Abstract
INTRODUCTION Hemophilia A is an inherited bleeding disorder due to a deficiency of coagulation factor VIII (FVIII). Prevention and treatment of bleeding is traditionally through intravenous infusion of a FVIII concentrate. Modifications of recombinant FVIII (rFVIII) with an aim to prolong the half-life have been modest, thought because FVIII is dependent on plasma von Willebrand factor (VWF) for its half-life. Efanesoctocog alfa (ALTUVIIIO), approved by the Federal Drug Administration (FDA) in February 2023, was made independent of endogenous VWF by linking of the FVIII-binding D'D3 domain of VWF to B-domain deleted single chain FVIII. AREAS COVERED This review will outline the development of efanesoctocog alfa and the pharmacokinetic and safety data from clinical trials, as well as efficacy data from the phase 3 trials. These data formed the basis for the FDA approval. EXPERT OPINION Efanesoctocog alfa is a new type of FVIII replacement with an extended half-life allowing once weekly dosing to achieve hemostasis and FVIII trough levels of 13-15 IU/dL. This provides a highly effective option for treatment and prevention of bleeding in hemophilia A, where FVIII levels are easily measured. It also provides an option for treatment of bleeding and coverage for surgery with few infusions.
Collapse
Affiliation(s)
- Barbara A Konkle
- Washington Center for Bleeding Disorders, Division of Hematology, University of Washington, Seattle, WA, USA
| |
Collapse
|
7
|
Swystun LL, Lillicrap D. Current Understanding of Inherited Modifiers of FVIII Pharmacokinetic Variation. Pharmgenomics Pers Med 2023; 16:239-252. [PMID: 36998673 PMCID: PMC10046206 DOI: 10.2147/pgpm.s383221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/06/2023] [Indexed: 04/01/2023] Open
Abstract
The inherited bleeding disorder hemophilia A involves the quantitative deficiency of the coagulation cofactor factor VIII (FVIII). Prophylactic treatment of severe hemophilia A patients with FVIII concentrates aims to reduce the frequency of spontaneous joint bleeding and requires personalized tailoring of dosing regimens to account for the substantial inter-individual variability of FVIII pharmacokinetics. The strong reproducibility of FVIII pharmacokinetic (PK) metrics between repeat analyses in the same individual suggests this trait is genetically regulated. While the influence of plasma von Willebrand factor antigen (VWF:Ag) levels, ABO blood group, and patient age on FVIII PK is well established, estimates suggest these factors account for less than 35% of the overall variability in FVIII PK. More recent studies have identified genetic determinants that modify FVIII clearance or half-life including VWF gene variants that impair VWF-FVIII binding resulting in the accelerated clearance of VWF-free FVIII. Additionally, variants in receptors that regulate the clearance of FVIII or the VWF-FVIII complex have been associated with FVIII PK. The characterization of genetic modifiers of FVIII PK will provide mechanistic insight into a subject of clinical significance and support the development of personalized treatment plans for patients with hemophilia A.
Collapse
Affiliation(s)
- Laura L Swystun
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| |
Collapse
|
8
|
Clinical Predictors and Prediction Models for rFVIII-Fc Half Life in Real-World People with Severe Hemophilia A. J Clin Med 2023; 12:jcm12062207. [PMID: 36983209 PMCID: PMC10053229 DOI: 10.3390/jcm12062207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/27/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
The half life of recombinant factor VIII-Fc (rFVIII-Fc) for people with hemophilia A (PwHA) varies greatly. Understanding the factors influencing the variation and assessment of rFVIII-Fc half life is important for personalized treatment. Eighty-five severe-type PwHA with rFVIII-Fc treatment receiving an evaluation of half life by the Web-Accessible Population Pharmacokinetic (PK) Service—Hemophilia during 2019–2021 were retrospectively enrolled. The 50-patient PK profiles before 2021 were used for analysis and developing prediction models of half life, and the 35-patient PK profiles in 2021 were used for external validation. The patients in the development cohort were aged 8–64, with a median rFVIII-Fc half life of 20.75 h (range, 8.25–41.5 h). By multivariate linear regression analysis, we found two, four, and five predictors of rFVIII-Fc half life for the blood groups non-O, O patients, and overall patients, respectively, including VWF:Ag, BMI, VWF:activity/VWF:Ag ratio, body weight, O blood group, inhibitor history, HCV infection, and hematocrit. The three prediction equations of rFVIII-Fc half life (T) were respectively developed as T for non-O group patients = −0.81 + 0.63 × (BMI, kg/m2) + 6.07 × (baseline VWF:Ag, IU/mL), T for O group patients = −0.68 + 13.30 × (baseline VWF:Ag, IU/mL) + 0.27 × (BW, kg) − 1.17 × (BMI, kg/m2) + 16.02 × (VWF:activity/VWF:Ag ratio), and T for overall patients = −1.76 + 7.24 × (baseline VWF:Ag, IU/mL) − 3.84 × (Inhibitor history) + 2.99 × (HCV infection) − 2.83 × (O blood group) + 0.30 × (Hct, %), which explained 51.97%, 75.17%, and 66.38% of the half life variability, respectively. For external validation, there was a significant correlation between the predicted and observed half lives in the validation cohort. The median half life deviation was +1.53 h, +1.28 h, and +1.79 h for the equations of non-O group, O group, and overall group patients, respectively. In total, eight predictors influencing rFVIII-Fc half life were identified. Prediction equations of rFVIII-Fc half life were developed for the non-O and O blood groups and overall PwHA with a good degree of external validation. The equations could be applied to patients aged 8–64 without the need for PK blood sampling and clinically valuable for personalized therapy.
Collapse
|
9
|
Elsheikh E, Lavin M, Heck LA, Larkin N, Mullaney B, Doherty D, Kennedy M, Keenan C, Guest T, O'Mahony B, Fazavana J, Fallon PG, Preston RJS, Gormley J, Ryan K, O'Connell NM, Singleton E, Byrne M, McGowan M, Roche S, Doyle M, Crowley MP, O'Shea SI, Reipert BM, Johnsen JM, Pipe SW, Di Paola J, Turecek PL, O'Donnell JS. Heterogeneity in the half-life of factor VIII concentrate in patients with hemophilia A is due to variability in the clearance of endogenous von Willebrand factor. JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH 2023; 21:1123-1134. [PMID: 36775768 DOI: 10.1016/j.jtha.2023.01.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/04/2023] [Accepted: 01/12/2023] [Indexed: 01/21/2023]
Abstract
BACKGROUND Previous studies have reported marked interindividual variation in factor VIII (FVIII) clearance in patients with hemophilia (PWH) and proposed a number of factors that influence this heterogeneity. OBJECTIVES To investigate the importance of the clearance rates of endogenous von Willebrand factor (VWF) compared with those of other FVIII half-life modifiers in adult PWH. METHODS The half-life of recombinant FVIII was determined in a cohort of 61 adult PWH. A range of reported modifiers of FVIII clearance was assessed (including plasma VWF:antigen and VWF propeptide levels; VWF-FVIII binding capacity; ABO blood group; and nonneutralizing anti-FVIII antibodies). The FVIII-binding region of the VWF gene was sequenced. Finally, the effects of variation in FVIII half-life on clinical phenotype were investigated. RESULTS We demonstrated that heterogeneity in the clearance of endogenous plasma VWF is a key determinant of variable FVIII half-life in PWH. Both ABO blood group and age significantly impact FVIII clearance. The effect of ABO blood group on FVIII half-life in PWH is modulated entirely through its effect on the clearance rates of endogenous VWF. In contrast, the age-related effect on FVIII clearance is, at least in part, VWF independent. In contrast to previous studies, no major effects of variation in VWF-FVIII binding affinity on FVIII clearance were observed. Although high-titer immunoglobulin G antibodies (≥1:80) were observed in 26% of PWH, these did not impact FVIII half-life. Importantly, the annual FVIII usage (IU/kg/y) was significantly (p = .0035) increased in patients with an FVIII half-life of <12 hours. CONCLUSION Our data demonstrate that heterogeneity in the half-life of FVIII concentrates in patients with hemophilia A is primarily attributable to variability in the clearance of endogenous VWF.
Collapse
Affiliation(s)
- Einas Elsheikh
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland; National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | - Michelle Lavin
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland; National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | - Lilian Antunes Heck
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Niamh Larkin
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | - Brendan Mullaney
- Haemostasis Molecular Diagnostics Laboratory, National Coagulation Centre, St. James's Hospital, Dublin, Ireland
| | - Dearbhla Doherty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Megan Kennedy
- Discipline of Physiotherapy, Trinity Centre for Health sciences, Trinity College Dublin, St James's Hospital, Dublin, Ireland
| | - Catriona Keenan
- Haemostasis Molecular Diagnostics Laboratory, National Coagulation Centre, St. James's Hospital, Dublin, Ireland
| | - Thomas Guest
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Judicael Fazavana
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Padraic G Fallon
- Inflammation and Immunity Research Group, Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - John Gormley
- Discipline of Physiotherapy, Trinity Centre for Health sciences, Trinity College Dublin, St James's Hospital, Dublin, Ireland
| | - Kevin Ryan
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | | | - Evelyn Singleton
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | - Mary Byrne
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | - Mark McGowan
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | - Sheila Roche
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | - Mairead Doyle
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | - Maeve P Crowley
- Department of Haematology, Cork University Hospital, Cork, Ireland
| | - Susan I O'Shea
- Department of Haematology, Cork University Hospital, Cork, Ireland
| | | | - Jill M Johnsen
- Bloodworks Northwest Research Institute, Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Steven W Pipe
- Departments of Pediatrics and Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jorge Di Paola
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Peter L Turecek
- Baxalta Innovations GmbH, A Member of the Takeda Group of Companies, Vienna, Austria
| | - James S O'Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland; National Coagulation Centre, St James's Hospital, Dublin, Ireland.
| |
Collapse
|
10
|
Ichinose A, Osaki T, Souri M, Favaloro EJ. A Review of Autoimmune Acquired von Willebrand Factor Deficiency in Japan. Semin Thromb Hemost 2022; 48:911-925. [PMID: 35803264 DOI: 10.1055/s-0042-1749088] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
von Willebrand factor (VWF) forms high-molecular-weight multimers and plays an essential role in hemostasis, and thus its deficiency leads to bleeding symptoms. Acquired von Willebrand syndrome (AVWS) is rare, but potentially underdiagnosed, and develops in various underlying disorders. AVWS caused by anti-VWF autoantibodies is a rare subcategory of AVWS that can also be referred to as autoimmune VWF deficiency (AiVWFD). We performed a search of patients with autoimmune coagulation factor deficiencies in our nationwide survey in Japan. Among these, suspected cases of AiVWFD were extremely few, with only 11 case consultations in the last 10 years. Of these, three and five were respectively positive for anti-VWF autoantibodies (anti-VWF-Ab) and VWF inhibitor (VWF-inh). We also performed an extensive literature search of other cases from Japan, and in total, 40 cases were finally identified to have AiVWFD, with mean age of 55.0 years. Most underlying disorders were lympho- or myeloproliferative diseases, followed by autoimmune diseases. The major bleeding sites were subcutaneous and mucosal, the bleeding severity was moderate, and there were no hemorrhagic deaths. Bleeding time was prolonged; factor VIII activity, VWF antigen, and VWF activity were decreased, and high-molecular-weight VWF multimers were absent or decreased. These are similar to the common abnormal laboratory findings observed among general AVWS cases. Hemostatic therapy often involved VWF concentrates and vasopressin, and antibody eradication therapy often included corticosteroids and achieved remission. Notably, of all cases, 68% had anti-VWF-Abs, and 83% of anti-VWF-Ab-positive patients were also VWF-inh positive. To accumulate precise clinical information on AiVWFD, it is necessary to verify and improve the measurement methods for both anti-VWF-Ab and anti-VWF-inh. These findings from Japan should be confirmed in other geographic localities.
Collapse
Affiliation(s)
- Akitada Ichinose
- Department of Molecular Patho-Biochemistry and Patho-Biology, Yamagata University School of Medicine, Yamagata, Japan.,The Japanese Collaborative Research Group (JCRG) on Autoimmune Acquired Coagulation Factor Deficiencies supported by the Japanese Ministry of Health, Labor and Welfare (MHLW), Yamagata, Japan
| | - Tsukasa Osaki
- Department of Molecular Patho-Biochemistry and Patho-Biology, Yamagata University School of Medicine, Yamagata, Japan.,The Japanese Collaborative Research Group (JCRG) on Autoimmune Acquired Coagulation Factor Deficiencies supported by the Japanese Ministry of Health, Labor and Welfare (MHLW), Yamagata, Japan.,Department of Public Health and Hygiene, Yamagata University Graduate School of Medical Science, Iida-Nishi, Yamagata, Japan
| | - Masayoshi Souri
- Department of Molecular Patho-Biochemistry and Patho-Biology, Yamagata University School of Medicine, Yamagata, Japan.,The Japanese Collaborative Research Group (JCRG) on Autoimmune Acquired Coagulation Factor Deficiencies supported by the Japanese Ministry of Health, Labor and Welfare (MHLW), Yamagata, Japan.,Department of Public Health and Hygiene, Yamagata University Graduate School of Medical Science, Iida-Nishi, Yamagata, Japan
| | - Emmanuel J Favaloro
- Department of Haematology, Sydney Centres for Thrombosis and Haemostasis, Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Westmead, New South Wales, Australia.,Faculty of Science and Health, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| |
Collapse
|
11
|
Predicting Individual Changes in Terminal Half-Life After Switching to Extended Half-Life Concentrates in Patients With Severe Hemophilia. Hemasphere 2022; 6:e694. [PMID: 35356797 PMCID: PMC8939912 DOI: 10.1097/hs9.0000000000000694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 01/29/2022] [Indexed: 12/02/2022] Open
Abstract
Predicting individual effects of switching from standard half-life (SHL) to extended half-life (EHL) FVIII/FIX concentrates is pivotal in clinical care, but large-scale individual data are scarce. The aim of this study was to assess individual changes in terminal half-life (THL) after switching to EHL concentrates and identifying determinants of a clinically relevant THL extension in people with severe hemophilia. Data from participants with pharmacokinetic studies on both SHL and EHL were extracted from the Web-Accessible Population Pharmacokinetics Service (WAPPS) database and stratified according to hemophilia type and age groups (children/adults). A 30% increase in THL was considered clinically relevant. Predictors of a relevant increase were identified using logistic regression. Data from 688 persons with severe hemophilia (2174 infusions) were included: 89% hemophilia A; median age: 21.7 (interquartile range [IQR]: 11.5–37.7); positive inhibitor history: 11.7%. THL increased by 38% (IQR: 17%–67%) and 212% (139%–367%) for hemophilia A and B, respectively. All EHL-FIX concentrate users showed clinically relevant THL extension. However, 40% (242/612) of people with hemophilia A showed limited extension or decrease in THL after switching. Relevant FVIII-THL extension was predicted by short baseline THL and blood group non-O in both children and adults. In conclusion, clinically relevant THL extension was observed in all 75/76 participants switching to EHL-FIX, and in 60% of 612 switching to EHL-FVIII. Short THL on SHL-FVIII and blood group non-O were identified as predictors for a relevant THL increase after switching to EHL-FVIII. Individualized pharmacokinetic assessment may guide clinical decision-making when switching from SHL to EHL-FVIII.
Collapse
|
12
|
Lim MY. How do we optimally utilize factor concentrates in persons with hemophilia? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:206-214. [PMID: 34889393 PMCID: PMC8791116 DOI: 10.1182/hematology.2021000310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The current mainstay of therapy for hemophilia is to replace the deficient clotting factor with the intravenous administration of exogenous clotting factor concentrates. Prophylaxis factor replacement therapy is now considered the standard of care in both pediatric and adult patients with hemophilia with a severe phenotype to protect musculoskeletal health and improve quality of life. Heterogeneity in bleeding presentation among patients with hemophilia due to genetic, environmental, and treatment-related factors has been well described. Accordingly, the World Federation of Hemophilia recommends an individualized prophylaxis regimen that considers the factors mentioned above to meet the clinical needs of the patient, which can vary over time. This review focuses on the practical points of choosing the type of factor concentrate, dose, and interval while evaluating appropriate target trough factor levels and bleeding triggers such as level of physical activity and joint status. We also discuss the use of a pharmacokinetics assessment and its incorporation in the clinic for a tailored approach toward individualized management. Overall, adopting an individualized prophylaxis regimen leads to an optimal utilization of factor concentrates with maximum efficacy and minimum waste.
Collapse
Affiliation(s)
- Ming Y. Lim
- Correspondence Ming Y. Lim, Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, 2000 Circle of Hope, Rm 4126, Salt Lake City, UT 84112; e-mail:
| |
Collapse
|
13
|
Lunghi B, Morfini M, Martinelli N, Balestra D, Linari S, Frusconi S, Branchini A, Cervellera CF, Marchetti G, Castaman G, Bernardi F. The Asialoglycoprotein Receptor Minor Subunit Gene Contributes to Pharmacokinetics of Factor VIII Concentrates in Hemophilia A. Thromb Haemost 2021; 122:715-725. [PMID: 34407556 DOI: 10.1055/a-1591-7869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
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. CONCLUSIONS 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.
Collapse
Affiliation(s)
- Barbara Lunghi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Massimo Morfini
- Italian Association Hemophilia Centers (AICE), Naples, Italy
| | | | - Dario Balestra
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Silvia Linari
- Department of Oncology, Center for Bleeding Disorders, Careggi University Hospital, Florence, Italy
| | - Sabrina Frusconi
- Genetic Diagnostics Unit, Laboratory Department, Careggi University Hospital, Florence, Italy
| | - Alessio Branchini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | | | - Giovanna Marchetti
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Giancarlo Castaman
- Department of Oncology, Center for Bleeding Disorders, Careggi University Hospital, Florence, Italy
| | - Francesco Bernardi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
14
|
Versloot O, Iserman E, Chelle P, Germini F, Edginton AN, Schutgens REG, Iorio A, Fischer K. Terminal half-life of FVIII and FIX according to age, blood group and concentrate type: Data from the WAPPS database. J Thromb Haemost 2021; 19:1896-1906. [PMID: 34013558 PMCID: PMC8361743 DOI: 10.1111/jth.15395] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND Real-life data on pharmacokinetics of factor (F) VIII/IX concentrates, especially extended half-life (EHL), concentrates in large cohorts of persons with hemophilia are currently lacking. OBJECTIVES This cross-sectional study aimed to establish reference values for terminal half-life (THL) for FVIII/IX concentrates according to concentrate type, age, blood group and inhibitor history. PATIENTS/METHODS Data were extracted from the Web-Accessible Population Pharmacokinetics Service database. Groups were compared by nonparametric tests. THL was modelled according to patient characteristics and concentrate type. RESULTS Infusion data (n = 8022) were collected from 4832 subjects (including 2222 children) with severe hemophilia (age: 1 month-85 years; 89% hemophilia A; 34% using EHL concentrates, 9.8% with history of inhibitors). THL of FVIII-EHL was longer than of FVIII standard half-life (SHL; median 15.1 vs. 11.1 h). FVIII-THL was dependent on age, concentrate type, blood group, and inhibitor history. THL of FIX-EHL was longer than of FIX-SHL (median 106.9 vs. 36.5 h). FIX-THL increased with age until 30 years and remained stable thereafter. FVIII-THL was shorter in subjects with blood group O. THL was decreased by 1.3 h for FVIII and 22 h for FIX in subjects with a positive inhibitor history. CONCLUSIONS We established reference values for FVIII/IX concentrates according to patient characteristics and concentrate type in a large database of hemophilia patients. These reference values may inform clinical practice (e.g., assessment of immune tolerance success), economic implications of procurement processes and value attribution of novel treatments (e.g., mimetics, gene therapy).
Collapse
Affiliation(s)
- Olav Versloot
- Van CreveldkliniekUniversity Medical Centre UtrechtUtrechtThe Netherlands
| | - Emma Iserman
- Department of Health Research Methods, Evidence, and ImpactMcMaster UniversityHamiltonONCanada
| | - Pierre Chelle
- School of PharmacyUniversity of WaterlooWaterlooONCanada
| | - Federico Germini
- Department of Health Research Methods, Evidence, and ImpactMcMaster UniversityHamiltonONCanada
- Department of MedicineMcMaster UniversityHamiltonONCanada
| | | | | | - Alfonso Iorio
- Department of Health Research Methods, Evidence, and ImpactMcMaster UniversityHamiltonONCanada
- Department of MedicineMcMaster UniversityHamiltonONCanada
- McMaster‐Bayer Endowed Research Chair in Clinical Epidemiology of Congenital bleeding DisordersDepartment of MedicineMcMasterHamiltonONCanada
| | - Kathelijn Fischer
- Van CreveldkliniekUniversity Medical Centre UtrechtUtrechtThe Netherlands
| | | |
Collapse
|
15
|
Goren R, Pullenayegum E, Blanchette VS, Dover S, Carcao M, Israels SJ, Chan A, Rivard GE, Steele M, Cloutier S, Klaassen RJ, Sinha R, Price VE, Laferriere N, Paradis E, Wu JK, Feldman BM. Patterns of joint damage in severe haemophilia A treated with prophylaxis. Haemophilia 2021; 27:666-673. [PMID: 34015166 DOI: 10.1111/hae.14345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 04/19/2021] [Accepted: 05/03/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The primary objective of this study was to assess whether there are different patterns (classes) of joint health in young boys with severe haemophilia A (SHA) prescribed primary tailored prophylaxis. We also assessed whether age at first index joint bleed, blood group, FVIII gene abnormality variant, factor VIII trough level, first-year bleeding rate and adherence to the prescribed prophylaxis regimen significantly predicted joint damage trajectory, and thus class membership. METHODS Using data collected prospectively as part of the Canadian Hemophilia Primary Prophylaxis Study (CHPS), we implemented a latent class growth mixture model technique to determine how many joint damage classes existed within the cohort. We used a multinomial logistic regression to predict the odds of class membership based on the above predictors. We fitted a survival model to assess whether there were differences in the rate of dose escalation across the groups. RESULTS We identified three distinct classes of trajectory: persistently low, moderately increasing and rapidly increasing joint scores. By multinomial regression, we found that only age at first index joint bleed predicted rapidly increasing joint scores. The rapidly increasing joint score class group moved through dose escalation significantly faster than the other two groups. CONCLUSIONS Using tailored prophylaxis, boys with SHA follow one of three joint health trajectories. By using knowledge of disease trajectories, clinicians may be able to adjust treatment according to a subject's predicted long-term joint health and institute cost-effective programmes of prophylaxis targeted at the individual subject level.
Collapse
Affiliation(s)
- Rachel Goren
- Institute of Health Policy, Management & Evaluation, The Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Eleanor Pullenayegum
- Institute of Health Policy, Management & Evaluation, The Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.,Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada
| | - Victor S Blanchette
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada.,Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Pediatrics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Saunya Dover
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada
| | - Manuel Carcao
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada.,Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Pediatrics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sara J Israels
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Anthony Chan
- Department of Pediatrics, McMaster Children's Hospital, McMaster University, Hamilton, ON, Canada
| | - Georges E Rivard
- Division of Hematology/Oncology, Department of Pediatrics, CHU Sainte-Justine, Montreal, QC, Canada
| | - MacGregor Steele
- Section of Pediatric Hematology, Department of Pediatrics, University of Calgary and Alberta Children's Hospital, Calgary, AB, Canada
| | - Stéphanie Cloutier
- Centre de l'hémophilie de l'est du Québec, Hôpital de l'Enfant-Jésus, Université Laval, Québec, QC, Canada
| | - Robert J Klaassen
- Division of Pediatric Hematology/Oncology, University of Ottawa, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Roona Sinha
- Saskatchewan Bleeding Disorders Program, Royal University Hospital, Saskatoon, SK, Canada
| | - Victoria E Price
- Division of Pediatric Hematology/Oncology, IWK Health Centre, Halifax, NS, Canada
| | - Nicole Laferriere
- Division of Hematology/Oncology, Thunder Bay Regional Cancer Care, Thunder Bay, ON, Canada
| | | | - John K Wu
- Division of Hematology/Oncology/BMT, Department of Pediatrics, UBC & BC Children's Hospital, Vancouver, BC, Canada
| | - Brian M Feldman
- Institute of Health Policy, Management & Evaluation, The Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.,Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Pediatrics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Division of Rheumatology, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
16
|
Di Minno A, Spadarella G, Esposito S, Mathew P, Di Minno G, Mannucci PM. Perspective - The case for zero bleeds and drug bioequivalence in the treatment of congenital hemophilia A in 2021. Blood Rev 2021; 50:100849. [PMID: 34024681 DOI: 10.1016/j.blre.2021.100849] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/13/2021] [Accepted: 05/04/2021] [Indexed: 01/19/2023]
Abstract
Not all patients with severe hemophilia A (HA) respond optimally to a given dose of a given product. Within-individual variance in cross-over studies makes each patient unique in the response to each standard half-life (SHL) factor VIII (FVIII) product in pharmacokinetic (PK) terms. This hampers the prediction of efficacy when a SHL FVIII product is employed. PK data showing that half-lives of SHL rFVIII are unsatisfactory to achieve zero bleeding in individual HA patients provide the rationale for switching from SHL to extended half-life (EHL) products. However, not all subjects receiving prophylaxis with EHL products achieve zero bleeding, the most cogent objective of personalized prophylaxis. Known determinants of FVIII half-life (age, von Willebrand factor [VWF] levels, blood group) cumulatively account for one third of the total inter-individual variation in FVIII clearance in subjects with severe HA. Investigations into precision, and accuracy of laboratory measurement to be employed; newer pathways for the clearance of both free-FVIII and VWF-bound FVIII, and adequately powered studies on omics and phenotypic heterogeneity, are likely to provide additional information on the remaining two thirds of inter-individual variation in FVIII clearance in HA. Variability in the clinical response has also been documented in patients when FVIII activity is mimicked by fixed subcutaneous doses of the bispecific antibody emicizumab. National registries that collect PK data of available FVIII products and ad hoc information on the individual response to emicizumab should be encouraged, to establish newer standards of care and ease personalized clinical decisions to achieve zero bleeding.
Collapse
Affiliation(s)
- Alessandro Di Minno
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II", Italy; CEINGE-Biotecnologie Avanzate, Università degli Studi di Napoli "Federico II", Italy.
| | - Gaia Spadarella
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", Italy
| | - Salvatore Esposito
- Dipartimento di Medicina Clinica e Chirurgia and Centro Hub per le Malattie Emorragiche Congenite e le Trombofilie, Università degli Studi di Napoli "Federico II", Italy
| | | | - Giovanni Di Minno
- Dipartimento di Medicina Clinica e Chirurgia and Centro Hub per le Malattie Emorragiche Congenite e le Trombofilie, Università degli Studi di Napoli "Federico II", Italy.
| | - Pier Mannuccio Mannucci
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy..
| |
Collapse
|
17
|
Rathaur VK, Vigneshwar NKV, Imran A, Pathania M, Agrawal S, Chacham S, Verma PK, Bhat NK. Rare but not Abdicated: Status of Haemophilia in foothills of Himalaya, Uttarakhand: A cross-sectional study. J Family Med Prim Care 2021; 10:1437-1442. [PMID: 34041191 PMCID: PMC8140229 DOI: 10.4103/jfmpc.jfmpc_1613_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/06/2020] [Accepted: 10/28/2020] [Indexed: 11/04/2022] Open
Abstract
Background Haemophilia is one of the bleeding disorders, which is inherited, in an xlinked recessive pattern. The diagnosis is by estimation of factor levels of 8 and 9. Timebound treatment for people living with Haemophilia (PWH) is factor replacement during bleeding manifestation. The prevalence of Haemophilia was mostly underestimated, and it is more so in hilly terrains like the state of Uttarakhand. Materials and Method This is a crosssectional study by compiling the data of PWH visiting the tertiary care centre for Haemophilia in Uttarakhand. We collected data from the patients with bleeding disorder reporting to the Haemophilia centre from July 2017 to December 2018. In this manuscript, we try to describe the pattern of Haemophilia and the degree of severity and incidence of inhibitors among the sample population of PWH who represent the population of Uttarakhand. The magnitude of problems faced by PWH from this hilly terrain to assess basic treatment in case of emergency is also being depicted. Result We reported Haemophilia A contributing about 80% of the PWH in our centre. Average distance a PWH has to travel to obtain treatment was about 131.5 km (SD ± 83.7 km). Incidence of inhibitors was about 5%. Conclusion We infer from our study that Hemophilia A is more common than Hemophilia B. Through this manuscript we hope to spread awareness of the Haemophilia care that is ongoing, the role of prophylaxis therapy and the future role of primary care physicians that may change the care of PWH in future.
Collapse
Affiliation(s)
- Vyas K Rathaur
- Professor and Head of the Department, Department of Pediatrics, Veer Chandra Singh Garhwali Govt. Institute of Medical Science & Research, N. K. V. Vigneshwar, Dehradun, Uttarakhand, India
| | - N K V Vigneshwar
- Junior Resident, Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Ayesha Imran
- Assistant Professor, Department of Pediatrics, Government Doon Medical College, Dehradun, Uttarakhand, India
| | - Monika Pathania
- Associate Professor, Department of Internal Medicine, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Sonam Agrawal
- Assistant Professor, Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Swathi Chacham
- Additional Professor, Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Prashant K Verma
- Associate Professor, Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Nowneet K Bhat
- Professor and Head of the Department, Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| |
Collapse
|
18
|
The relationship between ABO blood group, von Willebrand factor, and primary hemostasis. Blood 2021; 136:2864-2874. [PMID: 32785650 DOI: 10.1182/blood.2020005843] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/03/2020] [Indexed: 12/16/2022] Open
Abstract
Numerous studies have reported significant associations between ABO blood group and risk of cardiovascular disease. These studies have consistently demonstrated that thrombotic risk is significantly reduced in individuals in blood group O. Nevertheless, the biological mechanisms through which ABO influences hemostasis have remained poorly understood. Exciting recent data have provided novel insights into how these ABO effects are modulated and have highlighted that ABO group significantly influences platelet plug formation at sites of vascular injury (primary hemostasis). In particular, ABO affects multiple aspects of von Willebrand factor (VWF) biology. In keeping with their reduced thrombotic risk, plasma VWF levels are ∼25% lower in healthy group O compared with healthy group non-O individuals. In addition, blood group O VWF demonstrates enhanced susceptibility to ADAMTS13 proteolysis. Finally, preliminary findings suggest that the interaction of group O VWF with platelets may also be reduced. Although the molecular mechanisms underlying these ABO effects on VWF have not been fully elucidated, it seems likely that they are mediated in large part by the ABO(H) carbohydrate structures that are carried on both the N- and O-linked glycans of VWF. Interestingly, ABO(H) determinants are also expressed on several different platelet surface glycoprotein receptors. Recent studies support the hypothesis that ABO group not only exerts major quantitative and qualitative effects on VWF, but also affect specific aspects of platelet function. Given the severe morbidity and the mortality associated with thrombotic disorders, defining the mechanisms underlying these ABO effects is not only of scientific interest, but also of direct clinical importance.
Collapse
|
19
|
Ogiwara K, Swystun LL, Paine AS, Kepa S, Choi SJ, Rejtö J, Hopman W, Pabinger I, Lillicrap D. Factor VIII pharmacokinetics associates with genetic modifiers of VWF and FVIII clearance in an adult hemophilia A population. J Thromb Haemost 2021; 19:654-663. [PMID: 33219619 DOI: 10.1111/jth.15183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Factor VIII (FVIII) pharmacokinetics (PK) in adult hemophilia A populations are highly variable and have been previously determined to be influenced by von Willebrand factor:antigen (VWF:Ag), ABO blood group, and age. However, additional genetic determinants of FVIII PK are largely unknown. OBJECTIVES The contribution of VWF clearance, VWF-FVIII-binding activity, and genetic variants in VWF clearance receptors to FVIII PK in adult patients were assessed. METHODS FVIII PK assessment was performed in 44 adult subjects (age 18-61 years) with moderate or severe hemophilia A. VWF:Ag, VWF propeptide (VWFpp), VWFpp/VWF:Ag, and VWF:FVIII binding activity were measured. The VWF modifying loci CLEC4M, SCARA5, STAB2, and ABO, and the D'D3 FVIII-binding region of the VWF gene were genotyped. RESULTS VWF:Ag, VWFpp, and VWF:FVIIIB positively correlated with FVIII half-life and negatively correlated with FVIII clearance. VWFpp/VWF:Ag negatively correlated with FVIII half-life and positively correlated with FVIII clearance. The correlation between VWFpp/VWF:Ag and FVIII half-life was stronger for type non-O patients than for type O patients, suggesting that slower VWF clearance increases FVIII half-life. Patients heterozygous for the CLEC4M rs868875 variant had increased FVIII clearance when compared with individuals homozygous for the reference allele. The CLEC4M variable number of tandem repeat (VNTR) alleles were also associated with the rate of FVIII clearance. When compared with the quartile of patients with the fastest FVIII clearance, the quartile of patients with the slowest FVIII clearance had a decreased frequency of the CLEC4M 5-VNTR. CONCLUSIONS VWF-FVIII binding activity and genetic determinants of VWF clearance are important contributors to FVIII pharmacokinetics in adult patients.
Collapse
Affiliation(s)
- Kenichi Ogiwara
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Laura L Swystun
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - A Simonne Paine
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Sylvia Kepa
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Seon Jai Choi
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Judit Rejtö
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Wilma Hopman
- Department of Public Health Sciences, Queen's University, Kingston, ON, Canada
| | - Ingrid Pabinger
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| |
Collapse
|
20
|
Chen Z, Huang K, Li G, Zhen Y, Wu X, Di A, Liu G, Li Z, Alfonso I, Wu R. Pharmacokinetic variability of factor VIII concentrates in Chinese pediatric patients with moderate or severe hemophilia A. Pediatr Investig 2021; 5:38-45. [PMID: 33778426 PMCID: PMC7983998 DOI: 10.1002/ped4.12252] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/12/2020] [Indexed: 11/11/2022] Open
Abstract
IMPORTANCE The use of factor VIII (FVIII) concentrates under pharmacokinetic (PK) guidance has become the main approach for treatment of hemophilia. However, limited PK research has been conducted in Chinese pediatric patients. OBJECTIVE To investigate the PK parameters of various FVIII concentrates in Chinese pediatric patients. METHODS Seventy-nine patients were enrolled (28 treated with Kogenate FS®, 23 treated with Advate ®, and 28 treated with GreenMono™). All enrolled patients participated in single-dose PK analysis after at least a 3-day washout period. Blood samples were collected predose, as well as at 1 h, 9 h, 24 h, and 48 h after infusion; FVIII levels were measured using a one-stage clotting assay. von Willebrand Factor Antigen (VWF:Ag) levels and blood types were also determined. PK parameters were evaluated by WAPPS-Hemo. RESULTS Mean values of terminal elimination half-life time (t1/2) for the Kogenate FS®, Advate®, and GreenMono™ FVIII groups were 12.24 h, 10.18 h, and 9.62 h; median clearance values were 4.16, 6.23, and 5.11 mL·kg-1·h-1; and median in vivo recovery values were 1.97, 1.55, and 1.61 IU/dL per IU/kg. Longer t1/2, higher in vivo recovery, and lower clearance were observed in patients with higher VWF:Ag level who were treated with recombinant concentrates. INTERPRETATION Chinese pediatric patients with hemophilia had FVIII PK characteristics similar to those previously observed in non-Chinese children, including large variation among individuals. VWF:Ag level and FVIII brand were associated with differences in FVIII PK. Thus, PK-guided dosing should be used to optimize individualized therapy in Chinese children.
Collapse
Affiliation(s)
- Zhenping Chen
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of PediatricsMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Kun Huang
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of PediatricsMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Gang Li
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of PediatricsMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Yingzi Zhen
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of PediatricsMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Xinyi Wu
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of PediatricsMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Ai Di
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of PediatricsMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Guoqing Liu
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of PediatricsMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Zekun Li
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of PediatricsMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Iorio Alfonso
- Clinical Epidemiology and BiostatisticsMcMaster UniversityHamiltonONCanada
| | - Runhui Wu
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of PediatricsMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| |
Collapse
|
21
|
Franchini M, Coppola A, Santoro C, De Cristofaro R, Barillari G, Giordano P, Gresele P, Lassandro G, Napolitano M, Lucchini G, Luciani M, Ferretti A, Baldacci E, Riccardi F, Santoro RC, Pasca S, Zanon E, Tagliaferri A. ABO Blood Group and Inhibitor Risk in Severe Hemophilia A Patients: A Study from the Italian Association of Hemophilia Centers. Semin Thromb Hemost 2021; 47:84-89. [PMID: 33525041 DOI: 10.1055/s-0040-1718870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Considering the profound influence exerted by the ABO blood group system on hemostasis, mainly through the von Willebrand factor and factor VIII (FVIII) complex, we have conducted a study evaluating the possible role of blood type on the risk of inhibitor development in hemophilia A. A total of 287 consecutive Caucasian patients with severe hemophilia A (202 without FVIII inhibitors and 85 with FVIII inhibitors) followed at seven Italian Hemophilia Treatment Centers belonging to the Italian Association of Hemophilia Centers (AICE) were included in the study. A higher prevalence of O blood group was detected in patients without inhibitors as compared in inhibitor patients (55 vs. 30.6%; p < 0.001). Among the other variables analyzed (age, F8 mutation, type and intensity of treatment and treatment regimen), F8 mutation class (high-risk vs. low-risk), and treatment regimen (on-demand vs. prophylaxis) were significantly correlated with inhibitor development. However, on a multivariate analysis, only the effects of F8 mutation and ABO blood type were independent of other covariates, being that non-O blood type is associated with a 2.89-fold increased risk of inhibitor development. In conclusion, our study supports the protective effect of O blood type on inhibitor risk in severely affected hemophilia A patients.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantova, Italy
| | - Antonio Coppola
- Regional Reference Center for Inherited Bleeding Disorders, University Hospital, Parma, Italy
| | - Cristina Santoro
- Department of Hematology, Policlinico Umberto I University Hospital, Rome, Italy
| | - Raimondo De Cristofaro
- Hemorrhagic and Thrombotic Diseases Service, Area of Hematology, Fondazione Policlinico Universitario "A. Gemelli," IRCCS, Rome, Italy
| | - Giovanni Barillari
- Center for Hemorrhagic and Thrombotic Disorders, General and University Hospital, Udine, Italy
| | - Paola Giordano
- Pediatric Unit "B. Trambusti," Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro," Bari, Italy
| | - Paolo Gresele
- Section of Internal and Cardiovascular Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Giuseppe Lassandro
- Pediatric Unit "B. Trambusti," Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro," Bari, Italy
| | - Mariasanta Napolitano
- Hematology Unit, Department of Health Promotion, Mother and Child Care, Reference Regional Center for Thrombosis and Haemostasis, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Giuseppe Lucchini
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantova, Italy
| | - Matteo Luciani
- Oncohematology Department, Pediatric Hospital Bambino Gesù, Rome, Italy
| | - Antonietta Ferretti
- Department of Hematology, Policlinico Umberto I University Hospital, Rome, Italy
| | - Erminia Baldacci
- Department of Hematology, Policlinico Umberto I University Hospital, Rome, Italy
| | - Federica Riccardi
- Regional Reference Center for Inherited Bleeding Disorders, University Hospital, Parma, Italy
| | - Rita Carlotta Santoro
- Center for Hemorrhagic and Thrombotic Disorders, "Pugliese-Ciaccio" Hospital, Catanzaro, Italy
| | - Samantha Pasca
- Hemophilia Center, University Hospital of Padua, Padua, Italy
| | - Ezio Zanon
- Hemophilia Center, University Hospital of Padua, Padua, Italy
| | - Annarita Tagliaferri
- Regional Reference Center for Inherited Bleeding Disorders, University Hospital, Parma, Italy
| |
Collapse
|
22
|
Blanchette VS, Zunino L, Grassmann V, Barnes C, Carcao MD, Curtin J, Jackson S, Khoo L, Komrska V, Lillicrap D, Morfini M, Romanova G, Stephens D, Zapotocka E, Rand ML, Blatny J. A Practical, One-Clinic Visit Protocol for Pharmacokinetic Profile Generation with the ADVATE myPKFiT Dosing Tool in Severe Hemophilia A Subjects. Thromb Haemost 2021; 121:1326-1336. [PMID: 33506480 PMCID: PMC8494515 DOI: 10.1055/a-1376-0970] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Standard pharmacokinetic (PK) assessments are demanding for persons with hemophilia A, requiring a 72-hour washout and 5 to 11 timed blood samples. A no-washout, single-clinic visit, sparse sampling population PK (PPK) protocol is an attractive alternative. Here, we compared PK parameters obtained with a traditional washout, 6-sampling time point PPK protocol with a no-washout, single-clinic visit, reverse 2-sampling time point PPK protocol in persons with severe hemophilia A (SHA) receiving ADVATE. A total of 39 inhibitor-negative males with SHA (factor VIII activity [FVIII:C] < 2%) were enrolled in a prospective sequential design PK study. Participants completed a washout, 6-sampling time point PPK protocol as well as a no-washout, reverse 2-sampling time point protocol, with samples taken during a single 3-hour clinic visit 24 hours post home infusion of FVIII and then 3 hours post infusion in clinic. FVIII:C levels were analyzed by one-stage and chromogenic assays; blood group and von Willebrand factor antigen (VWF:Ag) were determined; and PK parameters were analyzed using the ADVATE myPKFiT dosing tool. There was moderate to almost perfect agreement for the PK parameters obtained with the 2- and the 6- point PPK protocols using a one-stage FVIII:C assay and a substantial to almost perfect agreement using a chromogenic FVIII:C assay. Significant associations between specific PK parameters and blood group and VWF:Ag were observed. The no-washout, single-clinic visit, reverse 2-sampling time point PPK protocol can be used in the routine clinical setting since it demonstrates sufficient accuracy compared with the more demanding and less practical washout, 6-sampling time point PPK protocol in persons with SHA receiving ADVATE.
Collapse
Affiliation(s)
- Victor S Blanchette
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Laura Zunino
- Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Viviane Grassmann
- Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Chris Barnes
- Haematology Department, The Royal Children's Hospital Melbourne, Victoria, Australia.,Haematology Research, Murdoch Children's Research Institute, Victoria, Australia
| | - Manuel D Carcao
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Julie Curtin
- Department of Haematology, The Children's Hospital at Westmead, Sydney, Australia.,Department of Paediatrics and Child Health, University of Sydney, Sydney, Australia
| | - Shannon Jackson
- Division of Haematology, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Liane Khoo
- Haematology Department, Royal Prince Alfred Hospital, NSW Health Pathology, Sydney, Australia
| | - Vladimir Komrska
- Department of Paediatric Haematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | | | - Gabriela Romanova
- Department of Clinical Haematology, University Hospital Brno, Brno, Czech Republic.,Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Derek Stephens
- Department of Clinical Research Services, The Hospital for Sick Children, Toronto, Canada
| | - Ester Zapotocka
- Department of Paediatric Haematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - Margaret L Rand
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Jan Blatny
- Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Paediatric Haematology, University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
23
|
Common Genetic Variants in ABO and CLEC4M Modulate the Pharmacokinetics of Recombinant FVIII in Severe Hemophilia A Patients. Thromb Haemost 2020; 120:1395-1406. [PMID: 32726853 DOI: 10.1055/s-0040-1714214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The pharmacokinetic (PK) response of severe hemophilia A (HA) patients to infused factor VIII (FVIII) shows substantial variability. Several environmental and genetic factors are associated with changes in FVIII plasma levels and infused FVIII PK. Based on the hypothesis that factors influencing endogenous FVIII can affect FVIII PK, the contribution of single-nucleotide variants (SNVs) in candidate genes was investigated in 51 severe HA patients. The effects of blood group, F8 variant type, von Willebrand factor antigen and activity levels, age, and weight were also explored. The myPKFiT device was used to estimate individual PK parameters, and SNVs and clinically reportable F8 variants were simultaneously analyzed in an Illumina MiSeq instrument, using the microfluidics-based Fluidigm Access Array system. The contribution of SNVs to FVIII half-life and clearance was addressed by robust regression modeling, taking into account other modulators. In line with previous studies, we provide robust evidence that age, body weight, and blood group, as well as SNVs in ABO and CLEC4M, participate in the variability of FVIII PK in HA patients. Main results: each copy of the rs7853989 (ABO) allele increases FVIII half-life by 1.4 hours (p = 0.0131) and decreases clearance by 0.5 mL/h/kg (p = 5.57E-03), whereas each additional rs868875 (CLEC4M) allele reduces FVIII half-life by 1.1 hours (p = 2.90E-05) and increases clearance by 0.3 mL/h/kg (p = 1.01E-03). These results contribute to advancing efforts to improve FVIII replacement therapies by adjusting to each patient's PK profile based on pharmacogenomic data. This personalized medicine will decrease the burden of treatment and maximize the benefits obtained.
Collapse
|
24
|
Turecek PL, Johnsen JM, Pipe SW, O'Donnell JS. Biological mechanisms underlying inter-individual variation in factor VIII clearance in haemophilia. Haemophilia 2020; 26:575-583. [PMID: 32596930 PMCID: PMC7496649 DOI: 10.1111/hae.14078] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/26/2020] [Indexed: 12/28/2022]
Abstract
Previous studies have highlighted marked inter‐individual variations in factor VIII (FVIII) clearance between patients with haemophilia (PWH). The half‐life of infused FVIII has been reported to vary from as little as 5.3 hours in some adult PWH, up to as long as 28.8 hours in other individuals. These differences in clearance kinetics have been consistently observed using a number of different plasma‐derived and recombinant FVIII products. Furthermore, recent studies have demonstrated that half‐life for extended half‐life (EHL‐) FVIII products also demonstrates significant inter‐patient variation. Since time spent with FVIII trough levels <1% has been shown to be associated with increased bleeding risk in PWH on prophylaxis therapy, this variability in FVIII clearance clearly has major clinical significance. Recent studies have provided significant novel insights into the cellular basis underlying FVIII clearance pathways. In addition, accumulating data have shown that endogenous plasma VWF levels, ABO blood group and age, all play important roles in regulating FVIII half‐life in PWH. Indeed, multiple regression analysis suggests that together these factors account for approximately 34% of the total inter‐individual variation in FVIII clearance observed between subjects with severe haemophilia A. In this review, we consider these and other putative modulators of FVIII half‐life, and discuss the biological mechanisms through which these factors impact upon FVIII clearance in vivo.
Collapse
Affiliation(s)
- Peter L Turecek
- Baxalta Innovations GmbH, A Member of the Takeda Group of Companies, Vienna, Austria
| | - Jill M Johnsen
- Bloodworks Northwest Research Institute, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Steven W Pipe
- Departments of Pediatrics and Pathology, University of Michigan, Ann Arbor, MI, USA
| | - James S O'Donnell
- Haemostasis Research Group, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland.,National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | | |
Collapse
|
25
|
Rayment R, Chalmers E, Forsyth K, Gooding R, Kelly AM, Shapiro S, Talks K, Tunstall O, Biss T. Guidelines on the use of prophylactic factor replacement for children and adults with Haemophilia A and B. Br J Haematol 2020; 190:684-695. [PMID: 32390158 DOI: 10.1111/bjh.16704] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/04/2020] [Accepted: 04/08/2020] [Indexed: 01/19/2023]
Affiliation(s)
| | | | | | | | - Anne M Kelly
- Cambridge University Hospitals NHS foundation Trust, Cambridge, UK
| | - Susan Shapiro
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Kate Talks
- The Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Oliver Tunstall
- University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Tina Biss
- The Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | | |
Collapse
|
26
|
Hajducek DM, Chelle P, Hermans C, Iorio A, McEneny-King A, Yu J, Edginton A. Development and evaluation of the population pharmacokinetic models for FVIII and FIX concentrates of the WAPPS-Hemo project. Haemophilia 2020; 26:384-400. [PMID: 32281726 DOI: 10.1111/hae.13977] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/14/2020] [Accepted: 03/05/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The Web-Accessible Population Pharmacokinetic Service (WAPPS) project generates individually predicted pharmacokinetic (PK) profiles and tailored prophylactic treatment regimens for haemophilic patients, which rely on a set of population PK (PopPK) models providing concentrate-specific priors for the Bayesian forecasting methodology. AIM To describe the predictive performance of the WAPPS PopPK models in use on the WAPPS-Hemo platform. METHODS Data for modelling include dense PK data obtained from industry sponsored and independent PK studies, and dense and sparse data accumulated through WAPPS-Hemo. WAPPS PopPK models were developed via non-linear mixed-effect modelling taking into account the effects of covariates and between-individual-and sometimes between-occasion-variability. Model evaluation consisted of (a) prediction-corrected Visual Predictive Check (pcVPC), (b) Limited Sampling Analysis (LSA) and (c) repeated hold-out cross-validation. RESULTS Thirty-three WAPPS PopPK models built on data from 3188 patients (ages 1-78 years) under treatment by factor VIII or IX products (FVIII, FIX) were evaluated. Overall, models exhibit excellent performance characteristics. The pcVPC shows that the observed PK data fall within acceptable 90% interpercentile predictive bands. A slight overprediction beyond the expected half-life, an anticipated result of using sparse data, occurs for some models. The LSA results in lower than 3% of relative error for FVIII and FIX products and 16% for engineered FIX products. Cross-Validation analysis yields relative errors lower than 1.5% and 1.4% in estimates of half-life and time to 0.02 IU/mL, respectively. CONCLUSION The WAPPS-Hemo models consistently showed excellent performance characteristics for the intended use for Bayesian forecasting of individual PK profiles.
Collapse
Affiliation(s)
| | - Pierre Chelle
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Cedric Hermans
- Haemostasis and Thrombosis Unit, Division of Haematology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Alfonso Iorio
- McMaster-Bayer Endowed Research Chair for Clinical Epidemiology of Congenital Bleeding Disorders, Department of Medicine, Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | | | - Jacky Yu
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Andrea Edginton
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
27
|
Genetic determinants of VWF clearance and FVIII binding modify FVIII pharmacokinetics in pediatric hemophilia A patients. Blood 2019; 134:880-891. [DOI: 10.1182/blood.2019000190] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Abstract
Factor VIII (FVIII) pharmacokinetic (PK) properties show high interpatient variability in hemophilia A patients. Although previous studies have determined that age, body mass index, von Willebrand factor antigen (VWF:Ag) levels, and ABO blood group status can influence FVIII PK, they do not account for all observed variability. In this study, we aim to describe the genetic determinants that modify the FVIII PK profile in a population of 43 pediatric hemophilia A patients. We observed that VWF:Ag and VWF propeptide (VWFpp)/VWF:Ag, but not VWFpp, were associated with FVIII half-life. VWFpp/VWF:Ag negatively correlated with FVIII half-life in patients with non-O blood type, but no correlation was observed for type O patients, suggesting that von Willebrand factor (VWF) half-life, as modified by the ABO blood group, is a strong regulator of FVIII PK. The FVIII-binding activity of VWF positively correlated with FVIII half-life, and the rare or low-frequency nonsynonymous VWF variants p.(Arg826Lys) and p.(Arg852Glu) were identified in patients with reduced VWF:FVIIIB but not VWF:Ag. Common variants at the VWF, CLEC4M, and STAB2 loci, which have been previously associated with plasma levels of VWF and FVIII, were associated with the FVIII PK profile. Together, these studies characterize the mechanistic basis by which VWF clearance and ABO glycosylation modify FVIII PK in a pediatric population. Moreover, this study is the first to identify non-VWF and non-ABO variants that modify FVIII PK in pediatric hemophilia A patients.
Collapse
|
28
|
Lunghi B, Bernardi F, Martinelli N, Frusconi S, Branchini A, Linari S, Marchetti G, Castaman G, Morfini M. Functional polymorphisms in the LDLR and pharmacokinetics of Factor VIII concentrates. J Thromb Haemost 2019; 17:1288-1296. [PMID: 31055871 DOI: 10.1111/jth.14473] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/29/2019] [Indexed: 01/04/2023]
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, β-coefficient -0.311 vs 0.348; CLD2, β-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.
Collapse
Affiliation(s)
- Barbara Lunghi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Francesco Bernardi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | | | - Sabrina Frusconi
- Genetic Diagnostics Unit, Laboratory Department, Careggi University Hospital, Florence, Italy
| | - Alessio Branchini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Silvia Linari
- Center for Bleeding Disorders, Department of Oncology, Careggi University Hospital, Florence, Italy
| | - Giovanna Marchetti
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Giancarlo Castaman
- Center for Bleeding Disorders, Department of Oncology, Careggi University Hospital, Florence, Italy
| | | |
Collapse
|
29
|
Loomans JI, Stokhuijzen E, Peters M, Fijnvandraat K. Administration of DDAVP did not improve the pharmacokinetics of FVIII concentrate in a clinically significant manner. J Clin Transl Res 2018; 3:351-357. [PMID: 30873482 PMCID: PMC6412613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The half-life and mean residence time (MRT) of infused recombinant factor VIII (FVIII) concentrate are associated with pre-infusion levels of von Willebrand factor (VWF) in severely affected hemophilia A patients. It is currently unknown if individual FVIII concentrate half-life and MRT can be extended by increasing endogenous VWF levels. Aim: Our aim was to evaluate the effect of a 1-deamino-8-D-arginine vasopressin (DDAVP)-induced rise in VWF concentration on the pharmacokinetics of infused FVIII in hemophilia A patients. METHODS Four adult hemophilia A patients participated in this cross-over, placebo-controlled study. Each patient received either intravenous DDAVP or placebo, one hour prior to administration of 50 IU/kg plasma-derived immune-affinity purified FVIII concentrate. RESULTS The combined administration of DDAVP and FVIII concentrate was well tolerated. The levels of VWF Antigen (Ag) doubled after DDAVP, whereas they remained stable after placebo infusion. This rise in VWF Ag resulted in a slight modification of the pharmacokinetic parameters of FVIII concentrate. The MRT of FVIII concentrate increased in all patients (mean from 17.6 h to 19.9 h, p < 0.001, 95% CI for MRT change: +4.7 to -0.3 h). However, in vivo recoveries tended to decrease following DDAVP administration. CONCLUSIONS Collectively, these data show that administration of DDAVP did not improve the pharmacokinetics of FVIII concentrate in a clinically significant manner. RELEVANCE FOR PATIENTS Our results indicate that no clinical benefit is to be expected from the modification in FVIII pharmacokinetics resulting from DDAVP-administration prior to infusion of FVIII concentrate in hemophilia A patients.
Collapse
Affiliation(s)
- Janneke I Loomans
- Emma Children's Hospital, Department of Pediatric Hematology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Eva Stokhuijzen
- Emma Children's Hospital, Department of Pediatric Hematology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Plasma Proteins, Sanquin Research, Amsterdam, the Netherlands
| | - Marjolein Peters
- Emma Children's Hospital, Department of Pediatric Hematology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Karin Fijnvandraat
- Emma Children's Hospital, Department of Pediatric Hematology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Plasma Proteins, Sanquin Research, Amsterdam, the Netherlands
| |
Collapse
|
30
|
Effect of ABO blood group on haemostatic parameters in severe haemophilia A patients performing acute moderate-intensity exercise. Blood Coagul Fibrinolysis 2018; 29:626-635. [PMID: 30036280 DOI: 10.1097/mbc.0000000000000762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
: The primary objective was to assess the effect of ABO blood group on von Willebrand factor (VWF) rise induced by four bouts of moderate-intensity physical activity, on pharmacokinetics of a B-domain-deleted recombinant FVIII (BDD-rFVIII), and haemostatic parameters in severe haemophilia A patients with a null mutation. The secondary objective was to compare the response to exercise according to infused product type in a subgroup of patients who previously participated to the same exercise protocol, while treated with a full length recombinant FVIII (FL-rFVIII). Twenty patients had two visits (rest and exercise). Blood samples were drawn before administration of BDD-rFVIII and at 6 time points, until 24 h postinfusion. FVIII activity increased transiently by 1.1-fold, but only after the first exercise session, as compared to rest. VWF:Ag and platelet count were significantly elevated after each session. Mean FVIII half-life and thromboelastography measurements were unchanged with exercise. However, 14 participants had a slight variation of FVIII half-life with exercise compared to rest (from -3.42 h to +2.51 h). Seven patients demonstrated a longer FVIII half-life (four with O blood group), whereas the remainders had a reduced half-life (three with O blood group). FVIII half-life correlated with baseline VWF:Ag at rest (r = 0.70, P < 0.001) and with exercise (r = 0.67, P < 0.002). Recovery was different between FL-rFVIII and BDD-rFVIII at rest (P = 0.032), but no significant differences were observed between half-life of products at rest and with exercise. ABO blood group and the type of rFVIII administered did not influence the response to exercise.
Collapse
|
31
|
Ibrahim UA, Ahmed SG. Determinants and modifiers of bleeding phenotypes in haemophilia-A: General and tropical perspectives. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2018. [DOI: 10.1016/j.ejmhg.2017.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
32
|
Franchini M, Mengoli C, Marano G, Pupella S, Mannucci PM, Liumbruno GM. The importance of ABO blood group in pharmacokinetic studies in haemophilia A. Haemophilia 2018; 24:e122-e123. [DOI: 10.1111/hae.13437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2018] [Indexed: 01/19/2023]
Affiliation(s)
- M. Franchini
- Department of Hematology and Transfusion Medicine; “Carlo Poma” Hospital; Mantua Italy
| | - C. Mengoli
- Italian National Blood Centre; National Institute of Health; Rome Italy
| | - G. Marano
- Italian National Blood Centre; National Institute of Health; Rome Italy
| | - S. Pupella
- Italian National Blood Centre; National Institute of Health; Rome Italy
| | - P. M. Mannucci
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center; Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico and University of Milan; Milan Italy
| | - G. M. Liumbruno
- Italian National Blood Centre; National Institute of Health; Rome Italy
| |
Collapse
|
33
|
Iorio A, Blanchette V, Blatny J, Collins P, Fischer K, Neufeld E. Estimating and interpreting the pharmacokinetic profiles of individual patients with hemophilia A or B using a population pharmacokinetic approach: communication from the SSC of the ISTH. J Thromb Haemost 2017; 15:2461-2465. [PMID: 29119666 DOI: 10.1111/jth.13867] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Indexed: 01/19/2023]
Affiliation(s)
- A Iorio
- Department of Health Research, Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - V Blanchette
- Division of Hematology/Oncology, Hospital for Sick Children and Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - J Blatny
- Department of Pediatric Hematology, University Hospital Brno, Brno, Czech Republic
| | - P Collins
- Arthur Bloom Haemophilia Centre, School of Medicine, University Hospital of Wales, Cardiff University, Cardiff, UK
| | - K Fischer
- Van Creveldkliniek, University Medical Center, Utrecht, the Netherlands
| | - E Neufeld
- St Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
34
|
Young PA, Migliorini M, Strickland DK. Evidence That Factor VIII Forms a Bivalent Complex with the Low Density Lipoprotein (LDL) Receptor-related Protein 1 (LRP1): IDENTIFICATION OF CLUSTER IV ON LRP1 AS THE MAJOR BINDING SITE. J Biol Chem 2016; 291:26035-26044. [PMID: 27794518 DOI: 10.1074/jbc.m116.754622] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/25/2016] [Indexed: 11/06/2022] Open
Abstract
Hemophilia A is a bleeding disorder caused by a deficiency in coagulation factor VIII (fVIII) that affects 1 in 5,000 males. Current prophylactic replacement therapy, although effective, is difficult to maintain due to the cost and frequency of injections. Hepatic clearance of fVIII is mediated by the LDL receptor-related protein 1 (LRP1), a member of the LDL receptor family. Although it is well established that fVIII binds LRP1, the molecular details of this interaction are unclear as most of the studies have been performed using fragments of fVIII and LRP1. In the current investigation, we examine the binding of intact fVIII to full-length LRP1 to gain insight into the molecular interaction. Chemical modification studies confirm the requirement for lysine residues in the interaction of fVIII with LRP1. Examination of the ionic strength dependence of the interaction of fVIII with LRP1 resulted in a Debye-Hückel plot with a slope of 1.8 ± 0.5, suggesting the involvement of two critical charged residues in the interaction of fVIII with LRP1. Kinetic studies utilizing surface plasmon resonance techniques reveal that the high affinity of fVIII for LRP1 results from avidity effects mediated by the interactions of two sites in fVIII with complementary sites on LRP1 to form a bivalent fVIII·LRP1 complex. Furthermore, although fVIII bound avidly to soluble forms of clusters II and IV from LRP1, only soluble cluster IV competed with the binding of fVIII to full-length LRP1, revealing that cluster IV represents the major fVIII binding site in LRP1.
Collapse
Affiliation(s)
- Patricia A Young
- From the Center for Vascular and Inflammatory Disease and the Departments of Surgery and Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Mary Migliorini
- From the Center for Vascular and Inflammatory Disease and the Departments of Surgery and Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Dudley K Strickland
- From the Center for Vascular and Inflammatory Disease and the Departments of Surgery and Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
35
|
Øie CI, Roepstorff K, Behrens C, Bøggild Kristensen J, Karpf DM, Bolt G, Gudme CN, Kjalke M, Smedsrød B, Appa RS. High-affinity von Willebrand factor binding does not affect the anatomical or hepatocellular distribution of factor VIII in rats. J Thromb Haemost 2016; 14:1803-13. [PMID: 27378673 DOI: 10.1111/jth.13406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Indexed: 11/29/2022]
Abstract
UNLABELLED Essentials Von Willebrand factor (VWF) stabilizes factor VIII (FVIII) and prevents its premature clearance. Rat anatomical and hepatocellular distribution studies assessed the VWF effect on FVIII clearance. Hepatocytes and liver sinusoidal endothelial cells play a key role in FVIII clearance. Anatomical and hepatocellular distribution of FVIII is independent of high-affinity VWF binding. ABSTRACT Background Von Willebrand factor (VWF) stabilizes factor VIII in the circulation and prevents its premature clearance. Objective To study the effects of VWF on FVIII clearance in rats with endogenous VWF. Methods Anatomical and hepatocellular distribution studies were performed in rats following intravenous administration of glycoiodinated recombinant FVIII (rFVIII) and a FVIII variant, FVIII-Y1680F, lacking high-affinity VWF binding. Radioactivity was quantified in organs, and in distinct liver cell populations. The role of VWF binding was also studied by immunohistochemical staining of rat livers perfused ex vivo with rFVIII alone or with a FVIII-binding VWF fragment. Results The liver was the predominant organ of rFVIII distribution, and a radioactivity peak was also observed in the intestines, suggesting FVIII secretion to the bile by hepatocytes. In the liver, ~60% of recovered radioactivity was associated with hepatocytes, 32% with liver sinusoidal endothelial cells (LSECs), and 9% with Kupffer cells (KCs). When calculated per cell, 1.5-fold to 3-fold more radioactivity was associated with LSECs than with hepatocytes. The importance of hepatocytes and LSECs was confirmed by immunohistochemical staining; strong staining was seen in LSECs, and less intense, punctate staining in hepatocytes. Minor staining in KCs was observed. Comparable anatomical and hepatocellular distributions were observed with rFVIII and FVIII-Y1680F, and the presence of the VWF fragment, D'D3A1, did not change the FVIII staining pattern in intact livers. Conclusions The present data support FVIII clearance via the liver, with hepatocytes and LSECs playing a key role. High-affinity VWF binding did not alter the anatomical or hepatocellular distribution of FVIII.
Collapse
Affiliation(s)
- C I Øie
- Vascular Biology Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - K Roepstorff
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - C Behrens
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | | | - D M Karpf
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - G Bolt
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - C N Gudme
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - M Kjalke
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - B Smedsrød
- Vascular Biology Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - R S Appa
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark.
| |
Collapse
|
36
|
McEneny-King A, Iorio A, Foster G, Edginton AN. The use of pharmacokinetics in dose individualization of factor VIII in the treatment of hemophilia A. Expert Opin Drug Metab Toxicol 2016; 12:1313-1321. [PMID: 27539370 DOI: 10.1080/17425255.2016.1214711] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Hemophilia A is a bleeding disorder resulting from a lack of clotting factor VIII (FVIII), and treatment typically consists of prophylactic replacement of the deficient factor. However, high between subject variability precludes the development of a 'one size fits all' dosing strategy and necessitates an individualized approach. We sought to summarize the data on the pharmacokinetics of FVIII available as a basis for the development of population pharmacokinetic models to be used in dose tailoring. Areas covered: We reviewed the pharmacokinetics of FVIII as used for the treatment of hemophilia A, with a focus on the variability observed between patients and the application of pharmacokinetic methods to dose individualization. We also explored the covariates affecting pharmacokinetic parameters, the differences between plasma-derived and recombinant FVIII and the development of extended half-life products. Expert opinion: The pharmacokinetics of factor VIII in patients with hemophilia shows a high interpatient variability, and is affected by age, weight, level of von Willebrand factor, and blood group. A population approach to estimating individual pharmacokinetics is likely to provide the most successful strategy to tailor factor concentrate dosing to the individual needs and to ensure optimal patient outcomes, while also improving the cost-effectiveness of prophylactic replacement therapy.
Collapse
Affiliation(s)
- Alanna McEneny-King
- a School of Pharmacy, Health Sciences Campus , University of Waterloo , Waterloo , ON , Canada
| | - Alfonso Iorio
- b Health Information Research Unit , McMaster University , Hamilton , ON , Canada
| | - Gary Foster
- c Clinical Epidemiology and Biostatistics , McMaster University , Hamilton , ON , Canada
| | - Andrea N Edginton
- a School of Pharmacy, Health Sciences Campus , University of Waterloo , Waterloo , ON , Canada
| |
Collapse
|
37
|
Zourikian N, Merlen C, Bonnefoy A, St-Louis J, Rivard GE. Effects of moderate-intensity physical exercise on pharmacokinetics of factor VIII and von Willebrand factor in young adults with severe haemophilia A: a pilot study. Haemophilia 2016; 22:e177-83. [PMID: 26988074 DOI: 10.1111/hae.12869] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2015] [Indexed: 11/30/2022]
Abstract
INTRODUCTION In persons with severe haemophilia A (pwshA), infused factor VIII (FVIII) half-life can vary according to such determinants as blood group, von Willebrand factor (VWF) level or age; however, FVIII pharmacokinetics (PK) has not been well studied in pwshA during exercise. AIM To investigate FVIII PK in pwshA performing moderate-intensity aerobic exercise. METHODS Twelve young-adult pwshA with the intron-22 inversion mutation, on relatively low-dose FVIII prophylaxis regimens, and relatively good musculoskeletal status were recruited. Abbreviated PK of FVIII activity and von Willebrand factor antigen (VWF:Ag) level were compared - during rest, and with 60-min exercise (2 × 15 min each of moderate-intensity stationary cycling and treadmill walking). During rest and exercise visits, a baseline blood specimen was drawn, routine prophylaxis FVIII infused; then six blood specimens were taken over the following 24 h. RESULTS For all subjects, mean half-life of infused FVIII did not change significantly with exercise vs. at rest (577 ± 190 vs. 614 ± 163 min; P = 0.4131). VWF:Ag rose transiently by 40-50% for 6-8 h with exercise (P < 0.01), particularly in non-O blood group subjects. No musculoskeletal bleeds occurred during the study. CONCLUSION Four × 15 min of moderate-intensity aerobic exercise increased VWF:Ag levels for 6-8 h, and showed no evidence of accelerated FVIII clearance or of musculoskeletal bleeding in these young-adult pwshA with relatively good musculoskeletal status, on relatively low-dose FVIII prophylaxis regimens. However, O blood group impact would merit larger studies, with longer durations of similar or more vigorous exercise intensities.
Collapse
Affiliation(s)
- N Zourikian
- Centre d'hémostase pédiatrique et adulte du CHU Sainte-Justine, Montréal, QC, Canada
| | - C Merlen
- Centre d'hémostase pédiatrique et adulte du CHU Sainte-Justine, Montréal, QC, Canada
| | - A Bonnefoy
- Centre d'hémostase pédiatrique et adulte du CHU Sainte-Justine, Montréal, QC, Canada
| | - J St-Louis
- Centre d'hémostase pédiatrique et adulte du CHU Sainte-Justine, Montréal, QC, Canada
| | - G E Rivard
- Centre d'hémostase pédiatrique et adulte du CHU Sainte-Justine, Montréal, QC, Canada
| |
Collapse
|
38
|
Henrard S, Hermans C. Impact of being overweight on factor VIII dosing in children with haemophilia A. Haemophilia 2015; 22:361-7. [PMID: 26558443 DOI: 10.1111/hae.12848] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 09/18/2015] [Accepted: 09/18/2015] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Treatment of haemophilia A (HA) requires infusions of factor VIII (FVIII) concentrates. The number of FVIII units infused to obtain a specific circulating FVIII level is calculated with the formula: [body weight (BW) (kg) × desired FVIII increase (%)]/2, with the assumption that each unit of FVIII infused per kg of BW increases the circulating FVIII level by 2%. AIM The aim of this study was to evaluate the impact of several morphometric parameters (BW, body mass index (BMI)-for-age, height), age and type of FVIII concentrate on FVIII recovery in children with HA. METHODS A total of 66 children aged between 10 and 18 with severe HA selected from six pharmacokinetic (PK) clinical trials using two recombinant FVIII concentrates were included in the analysis. Regression tree (RT) was used to identify predictors of FVIII recovery. RESULTS The median age was 14.5 years with a median FVIII recovery of 2.09 for all children. The median FVIII recovery was not significantly different between age groups. Two groups were created by RT: children with a BMI-for-age percentile <P95 (Median FVIII recovery: 1.94) and obese children with a BMI-for-age percentile ≥P95 (Median FVIII recovery: 2.65). The FVIII recovery was significantly different between these two groups (P < 0.001). CONCLUSION These results are consistent with previous studies conducted in adults with HA and confirm that the long-held and current practice of applying an arbitrary and universal recovery of two to the calculations of FVIII dosage should be abolished in both children and adults.
Collapse
Affiliation(s)
- S Henrard
- Institute of Health and Society (IRSS), Université catholique de Louvain, Brussels, Belgium.,Haemostasis and Thrombosis Unit, Division of Haematology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - C Hermans
- Haemostasis and Thrombosis Unit, Division of Haematology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
39
|
George LA, Camire RM. Profile of efraloctocog alfa and its potential in the treatment of hemophilia A. J Blood Med 2015; 6:131-41. [PMID: 25977610 PMCID: PMC4418388 DOI: 10.2147/jbm.s54632] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Hemophilia care has improved dramatically over the past 50 years, evolving from plasma concentrates, to purified plasma proteins, to recombinant clotting factors. These collective developments allowed for home delivery of on-demand and prophylactic treatment, resulting in the reduction of hemophilia morbidity and mortality and improved quality of life. Although efficacious in treating bleeding, conventional factor products’ half-lives require frequent venipuncture, which remains a significant burden to patients. Despite the remarkable advances in hemophilia care, no improvements have, until now, been made to the pharmacokinetic properties of factor products. Multiple strategies have more recently been employed to generate novel bioengineered products that, with great hope, represent the next wave of progress in hemophilia care. The use of these products will undoubtedly raise important discussion about choosing conventional factor over new long-acting factor products. Incorporation of these therapies into clinical care is accompanied by unanswered safety questions that will likely be evaluated only in postmarketing surveillance analysis. Further, these products may change current treatment paradigms with unclear cost repercussions and feasibility. This paper will review efraloctocog alfa (FVIII-Fc) and its role in the treatment of hemophilia A.
Collapse
Affiliation(s)
- Lindsey A George
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA ; Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rodney M Camire
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA ; Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA ; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
Optimal treatment strategies for hemophilia: achievements and limitations of current prophylactic regimens. Blood 2015; 125:2038-44. [DOI: 10.1182/blood-2015-01-528414] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Abstract
Prophylactic application of clotting factor concentrates is the basis of modern treatment of severe hemophilia A. In children, the early start of prophylaxis as primary or secondary prophylaxis has become the gold standard in most countries with adequate resources. In adults, prophylaxis is reasonably continued when started as primary or secondary prophylaxis in childhood to maintain healthy joint function. Initial data support that adult patients with already existing advanced joint arthropathy benefit from tertiary prophylaxis with significantly lowered number of bleeds, almost complete absence of target joints, and less time off from work. Current prophylactic regimens, although very effective, do not completely prevent joint disease in a long-term perspective. Joint arthropathy in primary prophylaxis develops over many years, sometimes over a decade or even longer time periods. The ankle joints are the first and most severely affected joints in those patients and thus may serve in outcome assessment as an indicator of early joint arthropathy when followed by ultrasound or magnetic resonance imaging. Optimized outcome and best use of available resources is expected from individualization of therapy regimens, which comprises the individual’s bleeding pattern, condition of the musculoskeletal system, level of physical activity and the pharmacokinetic profile of the substituted coagulation factor, and most recently includes novel products with extended half-lives.
Collapse
|
41
|
Mahdi AJ, Obaji SG, Collins PW. Role of enhanced half-life factor VIII and IX in the treatment of haemophilia. Br J Haematol 2015; 169:768-76. [PMID: 25754016 DOI: 10.1111/bjh.13360] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Treatment of congenital haemophilia with factor VIII and IX concentrates often requires frequent infusions. This has obvious implications in establishing effective administration strategies and, in turn, adherence. To overcome these issues, three main technologies--polyethylene-glycol, Fc-neonatal IgG1 and albumin fusion products--have emerged into various stages of clinical development. Published data indicates an approximately 1·5- and fivefold increase in half-life of factor VIII and IX, respectively, compared to standard recombinant concentrates. Studies into efficacy and safety are starting to be published. Monitoring and optimal use of these new concentrates remains unknown. Weekly factor IX prophylaxis appears to be a feasible prophylactic regimen in haemophilia B patients. Weekly longer-acting FVIII is unlikely to provide adequate prophylaxis in most patients with haemophilia A but may reduce the frequency of infusions. Ongoing clinical trials and real life experience will help shape how these products can be used in practice and their cost effectiveness. The drive for convenience however should not overshadow the ultimate goal of prophylaxis, namely, preventing bleeding and arthropathy.
Collapse
Affiliation(s)
- Ali J Mahdi
- Department of Haematology, University Hospital of Wales and School of Medicine Cardiff University, Cardiff, UK
| | - Samya G Obaji
- Department of Haematology, University Hospital of Wales and School of Medicine Cardiff University, Cardiff, UK
| | - Peter W Collins
- Department of Haematology, University Hospital of Wales and School of Medicine Cardiff University, Cardiff, UK
| |
Collapse
|
42
|
Abstract
To understand the placement of a certain protein in a physiological system and the pathogenesis of related disorders, it is not only of interest to determine its function but also important to describe the sequential steps in its life cycle, from synthesis to secretion and ultimately its clearance. von Willebrand factor (VWF) is a particularly intriguing case in this regard because of its important auxiliary roles (both intra- and extracellular) that implicate a wide range of other proteins: its presence is required for the formation and regulated release of endothelial storage organelles, the Weibel-Palade bodies (WPBs), whereas VWF is also a key determinant in the clearance of coagulation factor VIII. Thus, understanding the molecular and cellular basis of the VWF life cycle will help us gain insight into the pathogenesis of von Willebrand disease, design alternative treatment options to prolong the factor VIII half-life, and delineate the role of VWF and coresidents of the WPBs in the prothrombotic and proinflammatory response of endothelial cells. In this review, an update on our current knowledge on VWF biosynthesis, secretion, and clearance is provided and we will discuss how they can be affected by the presence of protein defects.
Collapse
|
43
|
Miesbach W, Berntorp E. Interaction between VWF and FVIII in treating VWD. Eur J Haematol 2015; 95:449-54. [DOI: 10.1111/ejh.12514] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2014] [Indexed: 11/26/2022]
Affiliation(s)
- Wolfgang Miesbach
- Medical Clinic III; Institute of Transfusion Medicine; Goethe University; Frankfurt Germany
| | - Erik Berntorp
- Centre for Thrombosis and Haemostasis; Skane University Hospital; Lund University; Malmö Sweden
| |
Collapse
|
44
|
Kepa S, Horvath B, Reitter-Pfoertner S, Schemper M, Quehenberger P, Grundbichler M, Heistinger M, Neumeister P, Mannhalter C, Pabinger I. Parameters influencing FVIII pharmacokinetics in patients with severe and moderate haemophilia A. Haemophilia 2015; 21:343-350. [DOI: 10.1111/hae.12592] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2014] [Indexed: 02/03/2023]
Affiliation(s)
- S. Kepa
- Clinical Division of Haematology and Haemostaseology; Department of Medicine I; Medical University of Vienna; Vienna Austria
| | - B. Horvath
- Department of Laboratory Medicine; Medical University of Vienna; Vienna Austria
| | - S. Reitter-Pfoertner
- Clinical Division of Haematology and Haemostaseology; Department of Medicine I; Medical University of Vienna; Vienna Austria
| | - M. Schemper
- Center for Medical Statistics, Informatics and Intelligent Systems; Medical University of Vienna; Vienna Austria
| | - P. Quehenberger
- Department of Laboratory Medicine; Medical University of Vienna; Vienna Austria
| | - M. Grundbichler
- Division of Haematology and Oncology; Department of Internal Medicine III; Medical University of Salzburg; Salzburg Austria
| | - M. Heistinger
- Department of Internal Medicine I; Clinical Centre of Klagenfurt am Woerthersee; Klagenfurt am Woerthersee Austria
| | - P. Neumeister
- Division of Haematology; Department of Internal Medicine; Medical University of Graz; Graz Austria
| | - C. Mannhalter
- Department of Laboratory Medicine; Medical University of Vienna; Vienna Austria
| | - I. Pabinger
- Clinical Division of Haematology and Haemostaseology; Department of Medicine I; Medical University of Vienna; Vienna Austria
| |
Collapse
|
45
|
Drury-Stewart DN, Lannert KW, Chung DW, Teramura GT, Zimring JC, Konkle BA, Gammill HS, Johnsen JM. Complex changes in von Willebrand factor-associated parameters are acquired during uncomplicated pregnancy. PLoS One 2014; 9:e112935. [PMID: 25409031 PMCID: PMC4237360 DOI: 10.1371/journal.pone.0112935] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/16/2014] [Indexed: 02/06/2023] Open
Abstract
Background The coagulation protein von Willebrand Factor (VWF) is known to be elevated in pregnancy. However, the timing and nature of changes in VWF and associated parameters throughout pregnancy are not well understood. Objectives To better understand the changes in VWF provoked by pregnancy, we studied VWF-associated parameters in samples collected over the course of healthy pregnancies. Methods We measured VWF antigen (VWF:Ag), VWF propeptide (VWFpp), Factor VIII (FVIII), and ADAMTS13 activity in samples collected from 46 women during pregnancy and at non-pregnant baseline. We also characterized pregnant vs. non-pregnant VWF multimer structure in 21 pregnancies, and performed isoelectric focusing (IEF) of VWF in two pregnancies which had samples from multiple trimesters. Results VWF:Ag and FVIII levels were significantly increased during pregnancy. ADAMTS13 activity was unchanged. VWFpp levels increased much later in pregnancy than VWF:Ag, resulting in a progressive decrease in VWFpp:Ag ratios. FVIII:VWF ratios also decreased in pregnancy. Most pregnancies exhibited a clear loss of larger VWF multimers and altered VWF triplet structure. Further evidence of acquired VWF qualitative changes in pregnancy was found in progressive, reversible shifts in VWF IEF patterns over gestation. Conclusions These data support a new view of pregnancy in which VWF can acquire qualitative changes associated with advancing gestational age. Modeling supports a scenario in which both increased VWF production and doubling of the VWF half-life would account for the data observed. We propose that gestation induces a prolongation in VWF survival, which likely contributes to increased total VWF levels and altered VWF structure.
Collapse
Affiliation(s)
| | - Kerry W. Lannert
- Research Institute, Puget Sound Blood Center, Seattle, Washington, United States of America
| | - Dominic W. Chung
- Research Institute, Puget Sound Blood Center, Seattle, Washington, United States of America
| | - Gayle T. Teramura
- Research Institute, Puget Sound Blood Center, Seattle, Washington, United States of America
| | - James C. Zimring
- Research Institute, Puget Sound Blood Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Barbara A. Konkle
- Research Institute, Puget Sound Blood Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Hilary S. Gammill
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jill M. Johnsen
- Research Institute, Puget Sound Blood Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
46
|
Carcao M. Changing paradigm of prophylaxis with longer acting factor concentrates. Haemophilia 2014; 20 Suppl 4:99-105. [DOI: 10.1111/hae.12405] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2014] [Indexed: 01/16/2023]
Affiliation(s)
- M. Carcao
- Division of Haematology/Oncology; Department of Pediatrics; Child Health Evaluative Sciences; Research Institute; The Hospital for Sick Children; Toronto ON Canada
| |
Collapse
|
47
|
Aledort L, Ljung R, Mann K, Pipe S. Factor VIII therapy for hemophilia A: current and future issues. Expert Rev Hematol 2014; 7:373-85. [PMID: 24717090 DOI: 10.1586/17474086.2014.899896] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Hemophilia A is a congenital, recessive, X-linked bleeding disorder that is managed with infusions of plasma-derived or recombinant factor (F) VIII. The primary considerations in FVIII replacement therapy today are the: 1) immunogenicity of FVIII concentrates, 2) role of longer-acting FVIII products, 3) prophylactic use of FVIII in children and adults with severe hemophilia A, and 4) affordability and availability of FVIII products. Improving patient outcomes by increasing the use of FVIII prophylaxis, preventing or eliminating FVIII inhibitors, and expanding access to FVIII concentrates in developing countries are the major challenges confronting clinicians who care for patients with hemophilia A.
Collapse
Affiliation(s)
- Louis Aledort
- Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1006, Newyork, NY, USA
| | | | | | | |
Collapse
|
48
|
Coyle TE, Reding MT, Lin JC, Michaels LA, Shah A, Powell J. Phase I study of BAY 94-9027, a PEGylated B-domain-deleted recombinant factor VIII with an extended half-life, in subjects with hemophilia A. J Thromb Haemost 2014; 12:488-96. [PMID: 24843882 PMCID: PMC4265842 DOI: 10.1111/jth.12506] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND BAY 94-9027 is a B-domain-deleted recombinant factor VIII (rFVIII) with site-specific attachment of poly(ethylene glycol) that has shown an extended half-life in animal models of hemophilia. OBJECTIVES To assess the pharmacokinetics and safety of BAY 94-9027 after single and repeated administration in subjects with severe hemophilia A. PATIENTS/METHODS This 8-week, prospective, multicenter, open-label, phase I trial was conducted in 14 subjects aged 21–58 years with FVIII of < 1%, ≥ 150 days of exposure to FVIII, and no history of FVIII inhibitors. After a ≥ 3-day washout, subjects received a single dose of sucrose-formulated rFVIII (rFVIII-FS) (cohort 1 [n = 7], 25 IU kg−1; cohort 2 [n = 7], 50 IU kg−1) for a 48-h pharmacokinetic (PK) study. After another ≥ 3-day washout, cohort 1 received twice-weekly BAY 94-9027 at 25 IU kg−1 (16 doses), and cohort 2 received once-weekly BAY 94-9027 at 60 IU kg−1 (nine doses). A 168-h PK study was performed after the first and last BAY 94-9027 doses. RESULTS BAY 94-9027 showed equivalent recovery and an improved PK profile vs. rFVIII-FS, with a half-life of ~ 19 h (vs. ~ 13.0 h for rFVIII-FS). BAY 94-9027 was well tolerated, and no immunogenicity was observed. CONCLUSIONS This phase I study demonstrates that BAY 94-9027 has an extended half-life in subjects with hemophilia A and, after multiple dosing, was well tolerated with no immunogenicity during the 8-week trial. A phase III study in a larger number of subjects is underway to fully characterize how this prolonged half-life will permit less frequent prophylaxis dosing for patients with hemophilia.
Collapse
|
49
|
Lalezari S, Martinowitz U, Windyga J, Enriquez MM, Delesen H, Schwartz L, Scharrer I. Correlation between endogenous VWF:Ag and PK parameters and bleeding frequency in severe haemophilia A subjects during three-times-weekly prophylaxis with rFVIII-FS. Haemophilia 2013; 20:e15-22. [PMID: 24252058 PMCID: PMC4233978 DOI: 10.1111/hae.12294] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2013] [Indexed: 11/30/2022]
Abstract
Patients with severe haemophilia A experience frequent and spontaneous bleeding, causing debilitating damage to joints and decreasing quality of life. Prophylaxis with factor VIII (FVIII) reduces joint damage if initiated early. Circulating FVIII levels may be influenced by endogenous von Willebrand factor (VWF), a chaperone protein that binds and stabilizes FVIII. The aim of this study was to determine whether endogenous VWF antigen (VWF:Ag) levels are correlated with FVIII pharmacokinetic (PK) parameters and clinical outcomes in patients with severe haemophilia A. Previously treated, non-inhibitor patients in a multinational, randomized, double-blind, Ph II study received prophylaxis with once-weekly BAY 79-4980 (35 IU kg−1) or thrice-weekly recombinant sucrose-formulated FVIII (rFVIII-FS; 25 IU kg−1). PK parameters were evaluated at weeks 1 and 26. The number of bleeds per patient during the study was captured as part of the core efficacy endpoint. Spearman rank correlations assessed relationships of VWF:Ag levels with patient age, PK and annualized bleeding rate. Of 131 study patients (aged 13−64 years; BAY 79-4980, n = 63; rFVIII-FS, n = 68), 27 (21%; n = 15 and 12 respectively) were evaluable for PK assessment. Baseline VWF:Ag levels correlated with patient age (P < 0.0001). There was no significant difference in PK results between treatments; thus, PK parameters and VWF levels of all patients were analysed together. AUCnorm and T1/2 significantly increased with increased VWF:Ag (P < 0.001); clearance significantly decreased with increased VWF:Ag (P = 0.002). Annualized bleeding rate in patients treated with 3× per week rFVIII-FS significantly correlated with VWF:Ag and age (P = 0.038 and 0.021 respectively). PK parameters as well as the clinical outcome significantly correlated with endogenous VWF:Ag. The improved clinical outcome in subjects with high VWF:Ag levels may be explained by VWF:Ag influence on FVIII PK.
Collapse
Affiliation(s)
- S Lalezari
- National Hemophilia Center, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | | | | | | | | | | | | |
Collapse
|
50
|
Shapiro A. Development of long-acting recombinant FVIII and FIX Fc fusion proteins for the management of hemophilia. Expert Opin Biol Ther 2013; 13:1287-97. [DOI: 10.1517/14712598.2013.819339] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|