1
|
Chowdary P, Carcao M, Kenet G, Pipe SW. Haemophilia. Lancet 2025; 405:736-750. [PMID: 40023652 DOI: 10.1016/s0140-6736(24)02139-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/27/2024] [Accepted: 09/26/2024] [Indexed: 03/04/2025]
Abstract
Haemophilia A and B are congenital X-linked bleeding disorders resulting from deficiencies in clotting factors VIII (haemophilia A) and IX (haemophilia B). Patients with severe deficiency, defined as having less than 1% of normal plasma factor activivity, often have spontaneous bleeding within the first few years of life. Those with moderate and mild deficiencies typically present with post-traumatic or post-surgical bleeding later in life. A high index of suspicion and measurement of factor activity in plasma facilitates early diagnosis. In the 21st century, therapeutic advances and comprehensive care have substantially improved both mortality and morbidity associated with these conditions. Management strategies for haemophilia include on-demand treatment for bleeding episodes and all surgeries and regular treatment (ie, prophylaxis) aimed at reducing bleeds, morbidity, and mortality, thereby enhancing quality of life. Treatment options include factor replacement therapy, non-replacement therapies that increase thrombin generation, and gene therapies that facilitate in vivo clotting factor synthesis. The therapies differ in their use for prophylaxis and on-demand treatment, the mode and frequency of administration, duration of treatment effect, degree of haemostatic protection, and side-effects. Monitoring the effectiveness of these prophylactic therapies involves assessing annual bleeding rates and joint damage. Personalised management strategies, which align treatment with individual goals (eg, playing competitive sports), initiated at diagnosis and maintained throughout the lifespan, are crucial for optimal outcomes. These strategies are facilitated by a multidisciplinary team and supported by clinician-led education for both clinicians and patients.
Collapse
Affiliation(s)
- Pratima Chowdary
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free Hospital, London, UK; Department of Haematology, Cancer Institute, University College London, London, UK.
| | - Manuel Carcao
- Department of Paediatrics, Division of Haematology/Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Gili Kenet
- National Haemophilia Center and The Amalia Biron Institute of Thrombosis & Hemostasis Research, Sheba Medical Center, Tel Aviv University, Ramat Gan, Israel
| | - Steven W Pipe
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
2
|
Mizoguchi Y, Hino M, Ueda H, Miyaguchi Y, Kobayashi M. Effectiveness of PK-Guided Personalized Recombinant FVIII Treatment in Patients with Hemophilia A: Clinical Case Experiences Based on an Observational Study. J Blood Med 2025; 16:27-39. [PMID: 39811825 PMCID: PMC11731019 DOI: 10.2147/jbm.s479564] [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: 05/23/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Purpose Prophylaxis with recombinant factor VIII (rFVIII) products is the gold-standard treatment for hemophilia A. However, interindividual differences affect the half-life and clearance of rFVIII products. The myPKFiT is a web-based medical-device software program for population pharmacokinetic (PK) simulation of FVIII products to guide accurate FVIII doses and dosing intervals. In this Japanese multicenter observational study, the efficacy of regimen adjustment using myPKFiT was examined. Patients and Methods Male patients with hemophilia A undergoing personalized treatment with myPKFiT using either octocog alfa or rurioctocog alfa pegol were included. Patients were aged <18 years. Primary endpoint was annualized bleeding rate (ABR). Secondary endpoints were ABR by type of bleeding, rFVIII product consumption, physical activity level, quality of life, and frequency of rFVIII administrations. Results are presented descriptively; however, for exploratory analysis, data before and after regimen adjustment were compared using the Wilcoxon signed-rank test. Results Seven patients aged 3-17 years (median age 13 years) participated in the study. Mean ABR for all bleeds decreased by 0.86 after PK-guided regimen adjustment. Four patients showed zero ABR before and after regimen adjustment using myPKFiT. No significant differences were noted in the consumption of rFVIII products. However, mean rFVIII consumption decreased in two patients after PK-guided regimen adjustment. Three patients increased physical activity and, according to the treatment based on the PK-guided regimen adjustment, this resulted in no increased bleeding. Conclusion The results from this study in a small number of patients suggest that PK-guided regimen adjustment with myPKFiT may support optimization of the individual prophylactic administration of the FVIII products octocog alfa and rurioctocog alfa pegol. Study Registration UMIN000044800.
Collapse
Affiliation(s)
- Yoko Mizoguchi
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Moeko Hino
- Department of Pediatrics, Chiba University Hospital, Chiba, Japan
| | - Hitoshi Ueda
- Japan Medical Office, Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | - Yasuo Miyaguchi
- Japan Medical Office, Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | - Masao Kobayashi
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
- Japanese Red Cross, Chugoku-Shikoku Block Blood Center, Hiroshima, Japan
| |
Collapse
|
3
|
Zhen Y, Ai D, Huang K, Li G, Chen Z, Wu R. The influence of dead space in blood sampling needle on FVIII level and pharmacokinetic profiles in children with hemophilia. Hematology 2024; 29:2314871. [PMID: 38346146 DOI: 10.1080/16078454.2024.2314871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 12/01/2023] [Indexed: 02/15/2024] Open
Abstract
OBJECTIVE To investigate the influence of the dead space in disposable blood sampling needle on activated partial thromboplastin time (APTT), FVIII level and pharmacokinetic (PK) profiles in children with hemophilia. METHODS Children (<18 years) with severe hemophilia A were enrolled. After three days' washout-period, blood samples were collected at pre-dose, 1 h, 3 h, 9 h, 24 h and 48 h post-infusion. At each timepoint, two 2 mL vacuum tubes with 3.2% trisodium citrate were used. The first tube was signed as 'non-standard' (NS) and the second tube was signed as 'standard' (S). FVIII activities were evaluated by one-stage assay. WAPPS-Hemo was used to generate PK profiles like half-life time (t1/2), clearance (CL), trough level and time to 1, 2 and 5IU/dL after a dose of 50 ± 10IU/dL. The FVIII activities at 9 h and 24 h post-infusion were put into WAPPS and thus brought four combinations by true or biased FVIII level that used. RESULT Compared with standard-collected blood samples, prolonged APTT results (P-values < 0.01) and decreased FVIII activity (P-values < 0.05) were revealed in those non-standard blood samples. The corresponding bias was in positive relation to both APTT-S (r = 0.44, P < 0.0001) and FVIII-S level(r = 0.68, P < 0.001). The FVIII bias percentage got larger as FVIII-S level reduced (r = -0.24, P < 0.01). During the four combinations of FVIII activity at 9 h and 24 h, statistically longer t1/2, lower CL and longer time to 1, 2 or 5IU/dL were observed in 9H-S&24H-S group and 9H-NS&24H-S group. CONCLUSION While using vacuum tubes for clotting indicators and PK profiles, the dead space of blood sampling needle should be eliminated in advance.
Collapse
Affiliation(s)
- Yingzi Zhen
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, People's Republic of China
| | - Di Ai
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, People's Republic of China
| | - Kun Huang
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, People's Republic of China
| | - Gang Li
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, People's Republic of China
| | - Zhenping Chen
- Hematologic Disease Laboratory, Hematology Center, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, People's Republic of China
| | - Runhui Wu
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, People's Republic of China
| |
Collapse
|
4
|
Aggarwal S, Tomar A, Kumar K, Kumar A, Kumar S, Garg S, Kumar N, Nandi S. The role of very low-dose prophylaxis in severe hemophilia patients. Indian J Pharmacol 2024; 56:312-316. [PMID: 39687953 PMCID: PMC11698291 DOI: 10.4103/ijp.ijp_333_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/31/2024] [Accepted: 10/24/2024] [Indexed: 12/18/2024] Open
Abstract
INTRODUCTION Very low-dose prophylaxis of antihemophilic factor (AHF) in hemophilia involves administration of minimal amounts of clotting factor concentrates, typically below standard doses, to maintain baseline levels and reduce bleeding episodes. This method uses smaller, more frequent doses to balance bleeding prevention and cost-effectiveness. Close monitoring ensures adequate protection while avoiding overtreatment, offering a tailored, cost-effective approach to improve the quality of life (QoL) of the hemophiliacs. AIM The study aims to study the bleeding risks in patients with severe hemophilia who are on very low-dose AHF prophylaxis. It also seeks to assess the annual consumption of AHF in the hemophiliacs. In addition, the research will determine the QoL in both groups using the social functioning (SF)-36 questionnaire,evaluate patient compliance with the prophylactic regimen, and check functional disability using the Functional Independence Score in Hemophilia (FISH). MATERIALS AND METHODS The patients selected for the study were diagnosed with cases of hemophilia and were planned for any surgical procedure. The study was a cross-sectional study lasting 12 months. Patients over 13 years were included and patients with any other bleeding tendencies were excluded from the study. RESULTS The differences found in the study, were significant in terms of annual bleed rate (ABR), amount of AHF consumed, SF-36, and FISH scores. CONCLUSION Our study found that very low-dose prophylaxis significantly reduced the ABR in hemophilia patients. Functional independence worsened in the control group but remained stable in the test group, which showed significant improvements in SF-36 domains. The test group had no inhibitor development and showed a decline in severe bleeds, proving the efficacy and superiority of low-dose prophylaxis over on-demand therapy.
Collapse
Affiliation(s)
- Sunita Aggarwal
- Department of Medicine, Lok Nayak Hospital, New Delhi, India
| | - Anshul Tomar
- Department of Medicine, Lok Nayak Hospital, New Delhi, India
| | - Kapil Kumar
- Department of Medicine, Lok Nayak Hospital, New Delhi, India
| | - Ankitesh Kumar
- Department of Medicine, Lok Nayak Hospital, New Delhi, India
| | - Suresh Kumar
- Department of Medicine, Lok Nayak Hospital, New Delhi, India
| | - Sandeep Garg
- Department of Medicine, Lok Nayak Hospital, New Delhi, India
| | - Naresh Kumar
- Department of Medicine, Lok Nayak Hospital, New Delhi, India
| | - Sudipta Nandi
- Department of Medicine, Lok Nayak Hospital, New Delhi, India
| |
Collapse
|
5
|
Meijon-Ortigueira MDM, Alvarez-Roman MT, De La Corte H, Butta N, Jimenez-Yuste V. Predicting joint involvement through tailored prophylaxis in severe haemophilia A, is it possible? Haemophilia 2024; 30:678-684. [PMID: 38575526 DOI: 10.1111/hae.15014] [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: 02/05/2024] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
INTRODUCTION Tailored prophylaxis is the current treatment regimen for patients with severe haemophilia A. Recently, published guidelines describe two possible approaches, based on clinical characteristics or estimation of pharmacokinetic parameters. However, both have strengths and weaknesses, and their characteristics need to be integrated to optimize treatment appropriately. In this paper, we present a model that considers together the characteristics of prophylaxis and the relevance of each. METHODS The age at initiation of prophylaxis, number of bleeding events, treatment regimen, therapeutic adherence, FVIII trough levels, and joint status were analyzed in 59 patients followed at La Paz University Hospital between January 2000 and December 2019. RESULTS The mean duration of primary prophylaxis of 113.37 ± 57.79 months. Eighty-three percent (n = 49) had no joint status involvement at the end of follow-up (HJHS and HEAD-US = 0). The median ABR was 0.7 (IQR 0.2 -1.0) and 54.2% presented trough levels of FVIII during follow-up >1 IU/dL. 72,9% engaged in some type of physical activity and overall adherence was over 85% in all patients evaluated. The regression analysis performed, considering all these factors, showed that the initiation of prophylaxis before 21 months of age was the most relevant protective factor against the appearance of joint involvement (OR 88.33 p.031 CI 95% 1.49-5224.40) CONCLUSION: Early initiation of prophylaxis was the most relevant factor in the protection of joint status. More comprehensive analysis models adapted to the characteristics of each population, are needed to adequately individualize treatment.
Collapse
Affiliation(s)
| | | | - Hortensia De La Corte
- Department of Physical Medicine and Rehabilitation, La Paz University Hospital-IdiPAZ, Madrid, Spain
| | - Nora Butta
- Department of Hematology, La Paz University Hospital-IdiPAZ, Madrid, Spain
| | - Victor Jimenez-Yuste
- Department of Hematology, La Paz University Hospital-IdiPAZ, Autonomous University of Madrid, Madrid, Spain
| |
Collapse
|
6
|
He H, Huang K, Cheng X, Wu X, Wu R, Wang X. Development and internal validation of a clinical prediction model for individualized dosing of BAY 81-8973, A full-length recombinant factor VIII, in pediatric patients with haemophilia A. Thromb Res 2023; 232:6-14. [PMID: 39492253 DOI: 10.1016/j.thromres.2023.10.009] [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: 07/18/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 11/05/2024]
Abstract
BACKGROUND As the most commonly used coagulation factor VIII (FVIII) concentrate in China, the individualized dosing prediction model of Kovaltry (BAY81-8973) is not fully investigated in pediatric patients. The prophylaxis tailored by population pharmacokinetic (PopPK) model can optimize dosing regimens. OBJECTIVES This study aimed to develop PopPK models of BAY 81-8973 in pediatric patients, identify quantitative relationships of blood type (as a substitution for von Willebrand factor) on FVIII clearance and provide model-informed precision dosing (MIPD) procedures. METHODS Pediatric patients with severe hemophilia A were enrolled and PK tests were conducted. The blood samples were collected at six time point. One-stage-based activated partial thromboplastin time was used for FVIII activity. Basic demographics and key covariates (blood type and von Willebrand factor antigen) were collected. A nonlinear mixed-effect modeling (NONMEM) approach was employed to establish PopPK model. Simulations were performed to evaluate current dosing regimens and present MIPD strategies. RESULTS A total of 30 pediatric patients were included in the analysis. In the final model, Fat-free mass calculated from weight, age, and height was included as a size descriptor which affect FVIII apparent volume of distribution and clearance. Both von Willebrand factor antigen (VWF:Ag) and blood type accounted for the interindividual variability of FVIII clearance, but only one can retain in the final model. Therefore, two PopPK models based on VWF:Ag or blood type were developed. When the VWF:Ag value is doubled, the FVIII clearance is reduced by 35 %. Compared with blood type non-O, the clearance in pediatric patients with blood type O increased by 25.9 %. Weight-based dosing without regard to age and blood type resulted in large differences in FVIII trough activity. Patient demographics, dosing information, sparse blood samples and PopPK model together with Bayesian estimate constituted the MIPD workflow. Using it, the individual parameters and optimized dosing regimen could be achieved. CONCLUSION This is the first predictive model designed to predict individualized dosing of BAY 81-8973 in pediatric patients with Hemophilia A. These results are useful in the PK-guided prophylaxis among pediatric patients and hold great potential to improve their long-term clinical outcomes.
Collapse
Affiliation(s)
- Huan He
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 10045, China
| | - Kun Huang
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 10045, China
| | - Xiaoling Cheng
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 10045, China
| | - Xinyi Wu
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 10045, China
| | - Runhui Wu
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 10045, China.
| | - Xiaoling Wang
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 10045, China.
| |
Collapse
|
7
|
Goedhart TMHJ, Janssen A, Mathôt RAA, Cnossen MH. The road to implementation of pharmacokinetic-guided dosing of factor replacement therapy in hemophilia and allied bleeding disorders. Identifying knowledge gaps by mapping barriers and facilitators. Blood Rev 2023; 61:101098. [PMID: 37321952 DOI: 10.1016/j.blre.2023.101098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 04/02/2023] [Accepted: 05/07/2023] [Indexed: 06/17/2023]
Abstract
Clinical guidelines and expert groups recommend the use of pharmacokinetic (PK)-guided dosing of factor replacement therapy for the treatment of bleeding disorders, especially for patients with hemophilia. Although PK-guided dosing is increasingly applied, it is generally not considered standard clinical practice. The aim of this scoping review is to map barriers and facilitators for the implementation of PK-guided dosing in clinical practice and to identify knowledge gaps. A literature search was performed and 110 articles were included that describe PK-guided dosing in patients with bleeding disorders, mostly hemophilia A. We defined two overarching themes, efficacy and feasibility, and discuss five topics within each theme. For each topic, barriers, facilitators and knowledge gaps were described. Although consensus was found with regard to some topics, contradicting reports were found for others, especially with respect to the efficacy of PK-guided dosing. These contradictions highlight the need for future research to elucidate current ambiguities.
Collapse
Affiliation(s)
- Tine M H J Goedhart
- Department of Pediatric Hematology and Oncology, Erasmus MC Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - A Janssen
- Department of Clinical Pharmacology - Hospital Pharmacy, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
| | - Ron A A Mathôt
- Department of Clinical Pharmacology - Hospital Pharmacy, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
| | - Marjon H Cnossen
- Department of Pediatric Hematology and Oncology, Erasmus MC Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
8
|
Bukkems LH, Jönsson S, Cnossen MH, Karlsson MO, Mathôt RAA. Relationship between factor VIII levels and bleeding for rFVIII-SingleChain in severe hemophilia A: A repeated time-to-event analysis. CPT Pharmacometrics Syst Pharmacol 2023; 12:706-718. [PMID: 36965157 DOI: 10.1002/psp4.12938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/31/2022] [Accepted: 01/31/2023] [Indexed: 03/27/2023] Open
Abstract
Publications on the exposure-effect relationships of factor concentrates for hemophilia treatment are limited, whereas such analyses give insight on treatment efficacy. Our objective was to examine the relationship between the dose, factor VIII (FVIII) levels and bleeding for rFVIII-SingleChain (lonoctocog alfa, Afstyla). Data from persons with severe hemophilia A on rFVIII-SingleChain prophylaxis from three clinical trials were combined. The published rFVIII-SingleChain population pharmacokinetic (PK) model was evaluated and expanded. The probability of bleeding was described with a parametric repeated time-to-event (RTTE) model. Data included 2080 bleeds, 2545 chromogenic stage assay, and 3052 one-stage assay FVIII levels from 241 persons (median age 19 years) followed for median 1090 days. The majority of the bleeds occurred in joints (65%) and the main bleeding reason was trauma (44%). The probability of bleeding decreased during follow-up and a FVIII level of 8.9 IU/dL (95% confidence interval: 6.9-10.9) decreased the bleeding hazard by 50% compared to a situation without FVIII in plasma. Variability in bleeding hazard between persons with similar FVIII levels was large, and the pre-study annual bleeding rate explained part of this variability. When a FVIII trough level of 1 or 3 IU/dL is targeted during prophylaxis, simulations predicted two (90% prediction interval [PI]: 0-17) or one (90% PI: 0-11) bleeds per year, respectively. In conclusion, the developed PK-RTTE model adequately described the relationship between dose, FVIII levels and bleeds for rFVIII-SingleChain. The obtained estimates were in agreement with those published for the FVIII concentrates BAY 81-8973 (octocog alfa) and BAY 94-9027 (damoctocog alfa pegol), indicating similar efficacy to reduce bleeding.
Collapse
Affiliation(s)
- Laura H Bukkems
- Hospital Pharmacy-Clinical Pharmacology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Siv Jönsson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Marjon H Cnossen
- Department of Pediatric Hematology, Erasmus University Medical Center - Sophia Children's Hospital Rotterdam, Rotterdam, The Netherlands
| | | | - Ron A A Mathôt
- Hospital Pharmacy-Clinical Pharmacology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
9
|
Persson S, Berndt C, Engstrand S, Trinczek A, Carlsson KS, Berntorp E. Area under the curve: Comparing the value of factor VIII replacement therapies in haemophilia A. Haemophilia 2023; 29:145-155. [PMID: 36445343 PMCID: PMC10098947 DOI: 10.1111/hae.14691] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION In factor VIII (FVIII) prophylaxis for haemophilia A, cost comparisons have used price per international unit (IU) based on the once reasonable assumption of equivalent outcome per IU. Now, with several extended half-life (EHL) products available, new outcome-oriented ways to compare products are needed. Area under the curve (AUC) quantifies FVIII levels over time after infusion providing comparable data. AIM To develop a decision analytical model for making indirect comparisons of FVIII replacement products based on AUC. METHODS A literature search identified 11 crossover studies with relevant pharmacokinetic data. A common comparator FVIII level curve was calculated using pooled data from selected studies. Absolute curves for other products were estimated based on relative differences to the common comparator (% difference vs the anchor). Three scenarios were investigated: (1) Kogenate® versus Kovaltry® and Jivi® ; (2) Advate® versus Elocta® , NovoEight® , Kovaltry, Adynovate® , Afstyla® , and ReFacto® ; and (3) Jivi versus Elocta, Adynovate, and Kogenate. Sensitivity analyses investigated effects of assay type and dose. RESULTS In scenario 1, Jivi (+50%) and Kovaltry (+14%) showed larger AUCs versus Kogenate. In scenario 2, EHL products, Elocta and Adynovate, had the largest AUC (+64% and +58%, respectively) versus Advate. Compared with all other products in scenario 3, Jivi had the largest AUC by +13%-28%. CONCLUSION This analysis concludes that EHL products differ in relative AUC, have a larger AUC compared with standard half-life, and thus, different FVIII levels over time after infusion. This model may aid decision makers in the absence of head-to-head data.
Collapse
Affiliation(s)
- Sofie Persson
- Swedish Institute for Health Economics, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Malmö, Sweden
| | | | | | | | - Katarina Steen Carlsson
- Swedish Institute for Health Economics, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Malmö, Sweden
| | - Erik Berntorp
- Department of Translational Medicine, Lund University, Lund, Malmö, Sweden
| |
Collapse
|
10
|
Urasiński T, Paczóska K, Badowska W, Bobrowska H, Dakowicz Ł, Dobaczewski G, Latos-Grażyńska E, Karolczyk G, Klukowska A, Kołtan A, Wojdalska M, Łaguna P, Niedźwiedzki M, Pietrys D, Radoń-Proskura J, Radwańska M, Rurańska I, Szczepański T, Wasiński D, Woźnica-Karczmarz I, Zielezińska K, Królak A, Ociepa T. Real pandemic world results of pharmacokinetic-tailored personalized prophylaxis of bleeds in Polish children and adolescents with severe hemophilia A. Front Pediatr 2023; 11:1084539. [PMID: 36911027 PMCID: PMC9996297 DOI: 10.3389/fped.2023.1084539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/08/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction In 2020, the new nationwide protocol of prophylaxis in Polish plasma-derived FVIII (pdFVIII) previously treated patients (PTPs) with severe hemophilia A (sHA) was introduced, resulting in the necessity of switching from pdFVIII to recombinant FVIII (octocog-alpha; rFVIII). The study aimed to: (1) assess the safety of switching from pdFVIII to rFVIII, (2) assess the safety and efficacy of pharmacokinetically based (PK-based) personalized prophylaxis in severe hemophilia A. Patients and methods 151 children and adolescents receiving prophylaxis with a standard dose (40 U/kg 3 x weekly) of pdFVIII were included in this study. Annualized bleeding rate (ABR) and annualized joint bleeding rate (AJBR) were analyzed for all patients before enrollment. Using myPKFiT application, pharmacokinetic (PK) analysis followed by the selection of the optimal model of prophylaxis was performed in all patients. Two possible models of prophylaxis (standard-dose rFVIII versus PK-based rFVIII) were discussed, with parents leaving the choice to their decision. Parents reported all episodes of bleeds. Screening for inhibitor was performed every 3 months. ABR and AJBR were prospectively analyzed again after a minimum follow-up time of 26 weeks. Results 141/151 (93.4%) patients completed the study. 34 patients decided to continue standard prophylaxis with rFVIII (Group I), whereas 107 were switched to PK-based prophylaxis (Group II). The risk of inhibitor development could be assessed in 137/151 (90.7%) patients. Only 2/137 (1.47%) patients (both on PK-based prophylaxis) developed low-titer inhibitor with its spontaneous elimination. The retrospective analysis of bleeds during the last 12 months of standard pdFVIII prophylaxis revealed that patients who decided to continue standard prophylaxis had historically lower ABR and AJBR than those who started PK-based personalized prophylaxis. After a minimum of 26 weeks, ABR and AJBR improved significantly in both groups. There was no significant difference in ABR and AJBR between Group I and Group II during the follow-up period. However, the rate of reduction of ABR and AJBR was higher in patients on PK-based personalized prophylaxis. Conclusion (1) Switching from pdFVIII to rFVIII (octocog-alpha) in PTPs with sHA is safe, (2) PK-based personalized prophylaxis may decrease ABR and AJBR in children and adolescents with sHA.
Collapse
Affiliation(s)
- Tomasz Urasiński
- Department of Pediatrics, Hemato-Oncology and Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Klaudia Paczóska
- Department of Pediatrics, Hemato-Oncology and Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Wanda Badowska
- Department of Pediatric Oncology and Hematology, School of Medicine, University of Warmia Mazury, Olsztyn, Poland
| | | | - Łucja Dakowicz
- Department of Pediatric Oncology and Hematology, Medical University of Białystok, Białystok, Poland
| | - Grzegorz Dobaczewski
- Department of Bone Marrow Transplantation, Pediatric Oncology and Hematology, Wrocław Medical University, Wrocław, Poland
| | - Elżbieta Latos-Grażyńska
- Department of Bone Marrow Transplantation, Pediatric Oncology and Hematology, Wrocław Medical University, Wrocław, Poland
| | - Grażyna Karolczyk
- II Department of Pediatrics, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| | - Anna Klukowska
- Department of Pediatric Oncology, Hematology and Transplantation, Medical University of Warsaw, Warszawa, Poland
| | - Andrzej Kołtan
- Department of Pediatrics, Hematology and Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Magdalena Wojdalska
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Łódź, Poland
| | - Paweł Łaguna
- Department of Pediatric Oncology, Hematology and Transplantation, Medical University of Warsaw, Warszawa, Poland
| | - Maciej Niedźwiedzki
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Danuta Pietrys
- Department of Pediatric Oncology and Hematology, Collegium Medicum, Jagiellonian University, Kraków, Poland
| | - Julia Radoń-Proskura
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Monika Radwańska
- Department of Pediatric Oncology and Hematology, College of Medical Sciences, University of Rzeszów, Rzeszów, Poland
| | - Iwona Rurańska
- Department of Pediatrics, Hematology and Oncology, Medical University of Silesia, Katowice, Poland
| | - Tomasz Szczepański
- Department of Pediatrics, Hematology and Oncology, Medical University of Silesia, Katowice, Poland
| | - Dariusz Wasiński
- Department of Pediatric Hematology and Oncology, Collegium Medicum, University of Zielona Góra, Zielona Góra, Poland
| | | | - Karolina Zielezińska
- Department of Pediatrics, Hemato-Oncology and Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Aleksandra Królak
- Department of Pediatrics, Hemato-Oncology and Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Tomasz Ociepa
- Department of Pediatrics, Hemato-Oncology and Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
11
|
Primacakti F, Sari TT, Gatot D, Sjakti HA, Chozie NA. Pharmacokinetic profile of children with haemophilia A receiving low-dose FVIII prophylaxis in Indonesia: A single centre experience. Haemophilia 2022; 28:720-725. [PMID: 35537097 DOI: 10.1111/hae.14584] [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: 09/14/2021] [Revised: 04/05/2022] [Accepted: 04/23/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Pharmacokinetic (PK) studies of low-dose prophylaxis (LDP) of coagulation factor VIII (FVIII) in children with severe haemophilia A (SHA) are scarce. OBJECTIVE This study aims to investigate the PK profile of children with SHA receiving LDP of FVIII. METHODS Paediatric patients receiving FVIII infusions (10 IU/kg twice weekly) were included. PK profiles were estimated using the Web Accessible Population Pharmacokinetic Service for Haemophilia (WAPPS-Haemo). The primary outcomes were the terminal half-life (t1/2 ), concentration-time profile, and time to reach an FVIII level of < 1%. The secondary outcome was the suggested dosing interval of FVIII prophylaxis based on the individual PK profile. RESULTS Twenty-five patients were recruited; their mean age was 12.3 ± 3.0 years. The t1/2 differed among patients receiving LDP of FVIII twice weekly, with a median of t1/2 was 14.8 h (IQR 12.6-16). The median time to reach an FVIII level of < 1% was 73.8 h (IQR 58.8-80.3). Most patients could maintain a trough level of FVIII > 1% longer than 48 h. At 72-96 h, patients needed a second dose of FVIII infusion because the FVIII level was < 1%. The suggested dosing interval of FVIII prophylaxis ranged from daily to every 96 h, depending on the individual PK profile. CONCLUSION Our study identified inter-individual differences in the PK parameters using LDP of FVIII twice weekly. The inter-individual results in different dosing intervals advise the timing of LDP. Estimating individual PK parameters enables the identification of the optimal prophylaxis frequency to prevent bleedings.
Collapse
Affiliation(s)
- Fitri Primacakti
- Paediatric Haematology-Oncology Division, Department of Child Health, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Teny T Sari
- Paediatric Haematology-Oncology Division, Department of Child Health, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Djajadiman Gatot
- Paediatric Haematology-Oncology Division, Department of Child Health, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Hikari A Sjakti
- Paediatric Haematology-Oncology Division, Department of Child Health, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Novie A Chozie
- Paediatric Haematology-Oncology Division, Department of Child Health, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
12
|
Goedhart TM, Bukkems LH, Moort I, Spence CC, Zwaan MC, Maat MP, Mathôt RA, Cnossen MH. Does difference between label and actual potency of factor VIII concentrate affect pharmacokinetic‐guided dosing of replacement therapy in haemophilia A? Haemophilia 2022; 28:610-618. [PMID: 35526235 PMCID: PMC9546314 DOI: 10.1111/hae.14575] [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: 09/23/2021] [Revised: 03/15/2022] [Accepted: 04/09/2022] [Indexed: 01/19/2023]
Abstract
Background Aim Methods Results Conclusion
Collapse
Affiliation(s)
- Tine M.H.J. Goedhart
- Department of Pediatric Hematology and Oncology Erasmus MC Sophia Children's Hospital University Medical Center Rotterdam Rotterdam The Netherlands
| | - Laura H. Bukkems
- Department of Clinical Pharmacology ‐ Hospital Pharmacy Amsterdam University Medical Centers Amsterdam The Netherlands
| | - Iris Moort
- Department of Hematology Erasmus MC University Medical Center Rotterdam Rotterdam The Netherlands
| | - Colin C. Spence
- Department of Pediatric Hematology and Oncology Erasmus MC Sophia Children's Hospital University Medical Center Rotterdam Rotterdam The Netherlands
| | - Michel C. Zwaan
- Department of Pediatric Hematology and Oncology Erasmus MC Sophia Children's Hospital University Medical Center Rotterdam Rotterdam The Netherlands
| | - Moniek P.M. Maat
- Department of Hematology Erasmus MC University Medical Center Rotterdam Rotterdam The Netherlands
| | - Ron A.A. Mathôt
- Department of Clinical Pharmacology ‐ Hospital Pharmacy Amsterdam University Medical Centers Amsterdam The Netherlands
| | - Marjon H. Cnossen
- Department of Pediatric Hematology and Oncology Erasmus MC Sophia Children's Hospital University Medical Center Rotterdam Rotterdam The Netherlands
| | | |
Collapse
|
13
|
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
|
14
|
Huang K, Wang Y, Zhen Y, Li G, Wu X, Zhang N, Chen Z, Wu R. Inter-individual variability in pharmacokinetics and clinical features in pediatric patients with severe hemophilia A. Thromb Res 2022; 213:71-77. [DOI: 10.1016/j.thromres.2022.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/02/2022] [Accepted: 03/09/2022] [Indexed: 01/13/2023]
|
15
|
Huang K, Zhen Y, Li G, Wu X, Chen Z, Wu R. Enhanced pharmacokinetics and reduced bleeds in boys with hemophilia A after switching to Kovaltry from other standard half-life factor VIII concentrates. Res Pract Thromb Haemost 2022; 6:e12686. [PMID: 35356665 PMCID: PMC8956787 DOI: 10.1002/rth2.12686] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 01/25/2023] Open
Abstract
Background BAY81-8973 (Kovaltry; Bayer, Berkeley, CA, USA) was reported with enhanced pharmacokinetic (PK) profiles compared with some other standard half-life (SHL) factor VIII (FVIII) concentrates. Limited head-to-head comparative studies were conducted in a real-world setting. Objective To make head-to-head comparisons of PK and clinical outcomes between Kovaltry and three other SHL FVIII concentrates. Methods Forty-seven boys with severe hemophilia A were enrolled and divided into three groups according to their previously used FVIII concentrates (Kogenate FS, N = 22; Advate, N = 14; GreenMono, N = 11). Two separate PK tests were conducted in each participant with a five-point assay during the study period from 6 months before switching to 6 months after switching. FVIII levels were detected by one-stage assay, and PK profiles were calculated by noncompartmental assay. Annualized bleeding rates were collected through participant' bleed logs. Results Longer half-life time (Kogenate FS group: 14.4 vs 11.9 hours, P < .0001; Advate group: 13.4 vs 9.7 hours, P < .0001; GreenMono group: 15.1 vs 10.7 hours, P < .001]) and lower clearance (Kogenate FS group: 3.3 vs 3.9 mL/kg/h, P < .01; Advate group: 3.7 vs 5.9 mL/kg/h, P < .01; GreenMono group: 3.0 vs 5.1 mL/kg/h, P < .01) were observed with Kovaltry. In addition, longer mean residential time (P < .01) and higher area under the curve (P < .01) were demonstrated. No statistical difference was found in in vivo recovery between Kovaltry and the other FVIII products. Participants who switched to Kovaltry from three other FVIII concentrates with the same dosing regimens obtained higher trough FVIII levels and better protection with reduced annualized bleeding rates. Conclusion Compared with Kogenate FS, Advate, and GreenMono, Kovaltry showed enhanced PK profiles, which contributed to reduced bleeding rates.
Collapse
Affiliation(s)
- Kun Huang
- Hematology CenterNational Center for Children's HealthBeijing Children’s HospitalCapital Medical UniversityBeijingChina
| | - Yingzi Zhen
- Hematology CenterNational Center for Children's HealthBeijing Children’s HospitalCapital Medical UniversityBeijingChina
| | - Gang Li
- Hematologic Disease LaboratoryBeijing Pediatric Research InstituteNational Center for Children's HealthBeijing Children's HospitalCapital Medical UniversityBeijingChina
| | - Xinyi Wu
- Hematology CenterNational Center for Children's HealthBeijing Children’s HospitalCapital Medical UniversityBeijingChina
| | - Zhenping Chen
- Hematologic Disease LaboratoryBeijing Pediatric Research InstituteNational Center for Children's HealthBeijing Children's HospitalCapital Medical UniversityBeijingChina
| | - Runhui Wu
- Hematology CenterNational Center for Children's HealthBeijing Children’s HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
16
|
Bukkems LH, Valke LLFG, Barteling W, Laros-van Gorkom BAP, Blijlevens NMA, Cnossen MH, van Heerde WL, Schols SEM, Mathôt RAA. Combining factor VIII levels and thrombin/plasmin generation: a population pharmacokinetic-pharmacodynamic model for patients with hemophilia A. Br J Clin Pharmacol 2021; 88:2757-2768. [PMID: 34921439 PMCID: PMC9304184 DOI: 10.1111/bcp.15185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/07/2021] [Accepted: 12/05/2021] [Indexed: 11/28/2022] Open
Abstract
Aims Prophylactic treatment of haemophilia A patients with factor VIII (FVIII) concentrate focuses on maintaining a minimal trough FVIII activity level to prevent bleeding. However, due to differences in bleeding tendency, the pharmacokinetic (PK)‐guided dosing approach may be suboptimal. An alternative approach could be the addition of haemostatic pharmacodynamic (PD) parameters, reflecting a patient's unique haemostatic balance. Our aim was to develop a population PK/PD model, based on FVIII activity levels and Nijmegen Haemostasis Assay (NHA) patterns, a global haemostatic assay that measures thrombin/plasmin generation simultaneously. Methods PK/PD measurements were collected from 30 patients treated with standard half‐life FVIII concentrate. The relationship between FVIII activity levels and the thrombin/plasmin generation parameters (thrombin potential, thrombin peak height and plasmin peak height), were described by sigmoidal Emax functions. Results The obtained EC50 value was smallest for the normalized thrombin potential (11.6 IU/dL), followed by normalized thrombin peak height (56.6 IU/dL) and normalized plasmin peak height (593 IU/dL), demonstrating that normalized thrombin potential showed 50% of the maximal effect at lower FVIII activity levels. Substantial inter‐individual variability in the PD parameters, such as EC50 of thrombin potential (86.9%) was observed, indicating that, despite similar FVIII activity levels, haemostatic capacity varies significantly between patients. Conclusion These data suggest that dosing based on patients' individual PK/PD parameters may be beneficial over dosing solely on individual PK parameters. This model could be used as proof‐of‐principle to examine the application of PK/PD‐guided dosing. However, the relation between the PD parameters and bleeding has to be better defined.
Collapse
Affiliation(s)
- Laura H Bukkems
- Department of Hospital Pharmacy-Clinical Pharmacology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Lars L F G Valke
- Department of Hematology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Hemophilia Treatment Centre, Nijmegen Eindhoven Maastricht, The Netherlands
| | - Wideke Barteling
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Britta A P Laros-van Gorkom
- Department of Hematology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Hemophilia Treatment Centre, Nijmegen Eindhoven Maastricht, The Netherlands
| | - Nicole M A Blijlevens
- Department of Hematology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Marjon H Cnossen
- Department of Pediatric Hematology, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Waander L van Heerde
- Hemophilia Treatment Centre, Nijmegen Eindhoven Maastricht, The Netherlands.,Enzyre BV, Novio Tech Campus, Nijmegen, The Netherlands
| | - Saskia E M Schols
- Department of Hematology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Hemophilia Treatment Centre, Nijmegen Eindhoven Maastricht, The Netherlands
| | - Ron A A Mathôt
- Department of Hospital Pharmacy-Clinical Pharmacology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
17
|
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
|
18
|
Lei B, Liang C, Feng H. Congenital hemophilia A with low activity of factor XII: a case report and literature review. Ital J Pediatr 2021; 47:204. [PMID: 34635150 PMCID: PMC8503991 DOI: 10.1186/s13052-021-01137-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/27/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Congenital hemophilia A is a recessive inherited hemorrhagic disorder. According to the activity of functional coagulation factors, the severity of hemophilia A is divided into three levels: mild, moderate and severe. The first bleeding episode in severe and moderate congenital hemophilia A occurs mostly in early childhood and mainly involves soft tissue and joint bleeds. At present, there are limited reports on severe congenital hemophilia A with low factor XII (FXII) activity during the neonatal period. CASE PRESENTATION A 13-day-old neonate was admitted to the hospital with hematoma near the joints of both upper arms. Coagulation tests showed he had low activity of factor VIII (FVIII) and FXII. He was diagnosed with congenital hemophilia A and treated with human coagulation factor VIII (recombinant FVIII). Although the hematoma became smaller, FVIII activity was only increased to a certain extent and FXII activity decreased gradually. Unfortunately, the child responded poorly to recombinant human coagulation factor VIII and his guardian rejected prophylactic inhibitors and genetic testing and refused further treatment. Three months later, the child developed intracranial hemorrhage (ICH) due to low FVIII activity. CONCLUSIONS In hemophilia A, the presence of FVIII inhibitors, drug concentration and testing are three important aspects that must be considered when FVIII activity does not reach the desired level. Early positive disease treatment and prophylaxis can decrease the frequency of bleeding and improve quality of life. We recommend that pregnant women with a family history of hemophilia A undergo early prenatal and neonatal genetic testing.
Collapse
Affiliation(s)
- Baoyu Lei
- Department of Neonatology, Maoming People's Hospital, Weimin Road, Maonan District, Maoming, 525000, Guangdong, China
| | - Chuang Liang
- Department of Neonatology, Maoming People's Hospital, Weimin Road, Maonan District, Maoming, 525000, Guangdong, China
| | - Haiyan Feng
- Department of Neonatology, Maoming People's Hospital, Weimin Road, Maonan District, Maoming, 525000, Guangdong, China.
| |
Collapse
|
19
|
Chelle P, Hajducek D, Mahdi M, Young S, Iorio A, Silvertown J, Edginton A. External qualification of the Web-Accessible Population Pharmacokinetic Service-Hemophilia (WAPPS-Hemo) models for octocog alfa using real patient data. Res Pract Thromb Haemost 2021; 5:e12599. [PMID: 34761154 PMCID: PMC8563921 DOI: 10.1002/rth2.12599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/23/2021] [Accepted: 09/02/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Existing adult patient pharmacokinetic (PK) data from the published Advate vs Kovaltry PK crossover study were used for this validation study. This data set is appropriate for qualification, given that it has not been previously submitted to Web-Accessible Population Pharmacokinetic Service-Hemophilia (WAPPS-Hemo) and will not have impacted the WAPPS-Hemo models for Kovaltry. OBJECTIVE To compare the population PK parameters for Kovaltry (BAY 81-8973) derived from the WAPPS-Hemo models with PK parameters derived from noncompartmental analysis (NCA), using a validation PK dataset. METHODS The qualification data set included Kovaltry factor activity (10 samples per infusion) and anthropometric data for 18 patients. Two analyses were performed comparison of Bayesian forecasting from the WAPPS-Hemo models versus NCA using the full 10-sample data set; and comparison of Bayesian forecasting using the full versus reduced 4- and 3-sample data sets. Agreement between outcomes was assessed by quantifying the variability and bias of the error. RESULTS Comparison of WAPPS-Hemo models versus NCA led to well-correlated outcomes despite a systematic overprediction of clearance. Population PK models demonstrated greater consistency with NCA on one-stage data, compared with chromogenic data. WAPPS-Hemo model results were consistent in reduced sampling compared to full sampling. Inclusion of a 48-hour time point in the reduced sampling greatly improved the consistency with full sampling. DISCUSSION Qualification of population PK models and their use for Bayesian forecasting in full and reduced sampling is an essential step toward their validation. The evaluations performed in this study support the confidence of PK parameter estimates provided by the models.
Collapse
Affiliation(s)
- Pierre Chelle
- School of PharmacyUniversity of WaterlooWaterlooONCanada
| | | | | | | | - Alfonso Iorio
- McMaster‐Bayer Endowed Research Chair for Clinical Epidemiology of Congenital Bleeding DisordersDepartment of MedicineDepartment of Health Research Methods, Evidence and ImpactMcMaster UniversityONCanada
| | | | | |
Collapse
|
20
|
Yoneyama K, Schmitt C, Chang T, Dhalluin C, Nagami S, Petry C, Levy GG. A Model-Based Framework to Inform the Dose Selection and Study Design of Emicizumab for Pediatric Patients With Hemophilia A. J Clin Pharmacol 2021; 62:232-244. [PMID: 34545950 PMCID: PMC9298840 DOI: 10.1002/jcph.1968] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/10/2021] [Indexed: 12/13/2022]
Abstract
Emicizumab is a bispecific antibody mimicking the cofactor function of activated coagulation factor VIII to prevent bleeds in patients with hemophilia A. The dose selection for the first-in-child phase III study of emicizumab was addressed by pediatric pharmacokinetic prediction using an adult/adolescent population pharmacokinetic model developed in phase I-I/II studies. The model was modified to incorporate functions describing the age-dependent increase in body weight (BW) with or without clearance maturation to account for the differences in emicizumab pharmacokinetics between adults/adolescents and children. A minimal dose anticipated to achieve in children the same target efficacious exposure as for adults/adolescents was identified when considering BW and clearance maturation. It was the same BW-based dose as for adults/adolescents and was selected for the starting dose for the pediatric study. Whether considering clearance maturation or not in addition to BW led to uncertainty in the pediatric pharmacokinetic prediction and dose selection, which informed implementation of a dose-adapting scheme in the study design. Exposure matching to adults/adolescents was ultimately achieved in children with the starting dose, indicating that consideration of clearance maturation in addition to BW provided adequate pediatric pharmacokinetic predictions for emicizumab. This pharmacokinetic finding in conjunction with exposure-response information served as a basis for the efficacy demonstrated in children, avoiding a time-consuming process for exploring an optimal pediatric dose of emicizumab. This experience indicates that a model-based framework helped optimize the pediatric dose selection and study design, thereby streamlining the development process with extrapolation, of emicizumab for children.
Collapse
Affiliation(s)
| | | | - Tiffany Chang
- Genentech, Inc., South San Francisco, California, USA
| | | | | | | | - Gallia G Levy
- Genentech, Inc., South San Francisco, California, USA
| |
Collapse
|
21
|
Zhu J, Wu YS, Beechinor RJ, Kemper R, Bukkems LH, Mathôt RAA, Cnossen MH, Gonzalez D, Chen SL, Key NS, Crona DJ. Pharmacokinetics of perioperative FVIII in adult patients with haemophilia A: An external validation and development of an alternative population pharmacokinetic model. Haemophilia 2021; 27:974-983. [PMID: 34405493 DOI: 10.1111/hae.14393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/04/2021] [Accepted: 07/25/2021] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Haemophilia A patients require perioperative clotting factor replacement to limit excessive bleeding. Weight-based dosing of Factor VIII (FVIII) does not account for inter-individual pharmacokinetic (PK) variability, and may lead to suboptimal FVIII exposure. AIM To perform an external validation of a previously developed population PK (popPK) model of perioperative FVIII in haemophilia A patients. METHODS A retrospective chart review identified perioperative haemophilia A patients at the University of North Carolina (UNC) between April 2014 and November 2019. Patient data was used to externally validate a previously published popPK model proposed by Hazendonk. Based on these validation results, a modified popPK model was developed to characterize FVIII PK in our patients. Dosing simulations were performed using this model to compare FVIII target attainment between intermittent bolus (IB) and continuous infusion (CI) administration methods. RESULTS A total of 521 FVIII concentrations, drawn from 34 patients, were analysed. Validation analyses revealed that the Hazendonk model did not fully capture FVIII PK in the UNC cohort. Therefore, a modified one-compartment model, with weight and age as covariates on clearance (CL), was developed. Dosing simulations revealed that CI resulted in improved target attainment by 16%, with reduced overall FVIII usage by 58 IU/kg, compared to IB. CONCLUSION External validation revealed a previously published popPK model of FVIII did not adequately characterize UNC patients, likely due to differences in patient populations. Future prospective studies are needed to evaluate our model prior to implementation into clinical practice.
Collapse
Affiliation(s)
- Jing Zhu
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Yi Shuan Wu
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Ryan J Beechinor
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA.,Department of Pharmacy, University of California Davis Comprehensive Cancer Center, Sacramento, California, USA
| | - Ryan Kemper
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Laura H Bukkems
- Hospital Pharmacy, Clinical Pharmacology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Ron A A Mathôt
- Hospital Pharmacy, Clinical Pharmacology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Marjon H Cnossen
- Department of Pediatric Hematology, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Sheh-Li Chen
- Department of Pharmacy, University of North Carolina Hospitals and Clinics, Chapel Hill, North Carolina, USA
| | - Nigel S Key
- Division of Hematology and Blood Research Center, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Daniel J Crona
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA.,Department of Pharmacy, University of North Carolina Hospitals and Clinics, Chapel Hill, North Carolina, USA.,UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
22
|
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
|
23
|
Megías-Vericat JE, Bonanad S, Haya S, Cid AR, Marqués MR, Ferrada A, Monte-Boquet E, Pérez-Alenda S, Bosch P, Querol-Giner F, Poveda JL. Clinical benefits of a Bayesian model for plasma-derived factor VIII/VWF after one year of pharmacokinetic-guided prophylaxis in severe/moderate hemophilia A patients. Thromb Res 2021; 205:99-105. [PMID: 34293540 DOI: 10.1016/j.thromres.2021.07.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/05/2021] [Accepted: 07/12/2021] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Individual pharmacokinetic (PK) profiling in hemophilia A (HA) helps to individualize prophylaxis using population PK models (popPK). A specific popPK model for plasma-derived factor VIII containing von-Willebrand Factor (pdFVIII/VWF) was developed. AIM To compare standard versus PK-driven prophylaxis, using a generic or a specific popPK model for pdFVIII/VWF. MATERIALS AND METHODS A prospective study conducted in HA patients in prophylaxis with pdFVIII/VWF (Fanhdi®) comparing three one-year study periods: (1) standard prophylaxis, (2) PK-guided prophylaxis using a generic pdFVIII popPK model which described FVIII activity irrespective of FVIII concentrate, and (3) PK-guided prophylaxis with specific pdFVIII/VWF popPK model. PK parameters analyzed were half-life, trough levels (TL) at 24, 48 and 72 h, and time to reach FVIII levels of 1, 2, 5% (T5%). Clinical outcomes were dose/kg, FVIII consumption, annualized bleeding rate (ABR), annualized joint bleeding rate (AJBR), spontaneous and traumatic bleeds. RESULTS Of the 30 analyzed patients, 28 had severe HA and the median age was 31.2. Fifteen patient's prophylaxis doses were PK-adjusted. After the generic PK-guided prophylaxis period, younger patients showed more joint bleeds, a shorter half-life, and lower TL48, TL72 and T5%. Using the specific pdFVIII/VWF popPK model compared with standard prophylaxis, a lower spontaneous AJBR was observed in the entire cohort and in patients aged >15 years. Additionally, lower spontaneous ABR was reported in patients aged ≤15 years comparing specific and generic models. CONCLUSIONS PK-guided prophylaxis with a specific pdFVIII/VWF popPK model allowed treatment individualization and improved bleeding control in routine clinical practice, especially in younger patients with short pdFVIII/VWF half-lives.
Collapse
Affiliation(s)
- Juan Eduardo Megías-Vericat
- Hospital Universitari i Politècnic La Fe, Pharmacy Department, Valencia, Spain; Hospital Universitari i Politècnic La Fe, Haemostasis and Thrombosis Unit, Valencia, Spain.
| | - Santiago Bonanad
- Hospital Universitari i Politècnic La Fe, Haemostasis and Thrombosis Unit, Valencia, Spain
| | - Saturnino Haya
- Hospital Universitari i Politècnic La Fe, Haemostasis and Thrombosis Unit, Valencia, Spain
| | - Ana Rosa Cid
- Hospital Universitari i Politècnic La Fe, Haemostasis and Thrombosis Unit, Valencia, Spain
| | | | - Alejandra Ferrada
- Hospital Universitari i Politècnic La Fe, Pharmacy Department, Valencia, Spain
| | - Emilio Monte-Boquet
- Hospital Universitari i Politècnic La Fe, Pharmacy Department, Valencia, Spain
| | - Sofía Pérez-Alenda
- Physiotherapy in Motion Multispeciality Research Group (PTinMOTION), Department of Physiotherapy, University of Valencia
| | - Pau Bosch
- Hospital Universitari i Politècnic La Fe, Haemostasis and Thrombosis Unit, Valencia, Spain
| | - Felipe Querol-Giner
- Physiotherapy in Motion Multispeciality Research Group (PTinMOTION), Department of Physiotherapy, University of Valencia
| | - Jose Luis Poveda
- Hospital Universitari i Politècnic La Fe, Pharmacy Department, Valencia, Spain
| |
Collapse
|
24
|
Jonsson F, Schmitt C, Petry C, Mercier F, Frey N, Retout S. Exposure-Bleeding Count Modeling of Emicizumab for the Prophylaxis of Bleeding in Persons with Hemophilia A with/Without Inhibitors Against Factor VIII. Clin Pharmacokinet 2021; 60:931-941. [PMID: 33709296 PMCID: PMC8249270 DOI: 10.1007/s40262-021-01006-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND OBJECTIVE Emicizumab is a monoclonal antibody that bridges activated coagulation factor IX and factor X to restore effective hemostasis in persons with hemophilia A. It is indicated for routine prophylaxis of bleeding episodes in persons with hemophilia A. The aim of the present study is to describe the exposure-response relationship between emicizumab concentrations and bleeding frequency, and to confirm adequate bleeding control of the investigated dosing regimens 1.5 mg/kg once weekly, 3 mg/kg every 2 weeks, and 6 mg/kg every 4 weeks. METHODS Treated bleeding events were pooled from 445 persons with hemophilia A with and without inhibitors against factor VIII, participating in six clinical studies. Emicizumab concentrations were predicted using a previously developed population pharmacokinetic model. A count model was used to quantify the exposure-response relationship. These models were used to illustrate the relationship between emicizumab concentrations and cumulative count of bleeding over 1 year (annualized bleeding rate). RESULTS The final exposure-response model, based on a generalized Poisson distribution and an inhibitory Emax relationship, adequately describes the relationship between daily emicizumab concentrations and daily bleed frequency. A significant effect of factor VIII prophylaxis among persons with hemophilia A without inhibitors was found. Annualized bleeding rate simulations show that the three emicizumab dosing regimens maintain the concentrations close to the plateau of the effect. At the average steady-state concentration across all regimens (53.5 µg/mL), the predicted mean annualized bleeding rate is 1.28, corresponding to a 94.0% reduction from baseline. CONCLUSIONS These results confirm that average emicizumab concentrations achieved with all three emicizumab dosing regimens provide adequate bleeding control.
Collapse
Affiliation(s)
| | - Christophe Schmitt
- Clinical Pharmacology, Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Claire Petry
- Clinical Pharmacology, Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Francois Mercier
- Clinical Pharmacology, Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Nicolas Frey
- Clinical Pharmacology, Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Sylvie Retout
- Clinical Pharmacology, Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| |
Collapse
|
25
|
Abstract
Haemophilia A and B are rare congenital, recessive X-linked disorders caused by lack or deficiency of clotting factor VIII (FVIII) or IX (FIX), respectively. The severity of the disease depends on the reduction of levels of FVIII or FIX, which are determined by the type of the causative mutation in the genes encoding the factors (F8 and F9, respectively). The hallmark clinical characteristic, especially in untreated severe forms, is bleeding (spontaneous or after trauma) into major joints such as ankles, knees and elbows, which can result in the development of arthropathy. Intracranial bleeds and bleeds into internal organs may be life-threatening. The median life expectancy was ~30 years until the 1960s, but improved understanding of the disorder and development of efficacious therapy based on prophylactic replacement of the missing factor has caused a paradigm shift, and today individuals with haemophilia can look forward to a virtually normal life expectancy and quality of life. Nevertheless, the potential development of inhibitory antibodies to infused factor is still a major hurdle to overcome in a substantial proportion of patients. Finally, gene therapy for both types of haemophilia has progressed remarkably and could soon become a reality.
Collapse
|
26
|
Astermark J, Olsson A, Chelle P, Täckström K, Walger M, Magnusson M, Iorio A. Comparison of single subject and population-based pharmacokinetics for optimizing prophylaxis with simoctocog alfa in patients with haemophilia A. Haemophilia 2021; 27:626-633. [PMID: 33966319 DOI: 10.1111/hae.14329] [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: 12/18/2020] [Revised: 03/30/2021] [Accepted: 04/21/2021] [Indexed: 01/19/2023]
Abstract
INTRODUCTION The use of pharmacokinetic assessment for optimal prophylactic dosing of factor concentrates in haemophilia has gained increasing enthusiasm over the last decade. However, blood sampling on several occasions is burdensome and limited sampling using population-based PK is appealing. AIM To compare the pharmacokinetics and dosing recommendations for prophylaxis using six-point single subject versus population-based method (WAPPS-Hemo) for simoctocog alfa (Nuwiq® ). METHODS Twelve adult patients with severe haemophilia A received a factor VIII (FVIII) dose of ≈50 IU/kg, and the activity was measured pre-infusion and at 30 min, 6, 9, 24 and 48 h post-infusion. Half-life (t1 /2 ), weight-normalized AUC and time to troughs of 5%, 3% and 1% were calculated. The correlation between the PK algorithms was assessed using intraclass correlations (ICC) and dosing estimations were provided. RESULTS WAPPS-Hemo yielded a slightly longer mean t1 /2 , but the overall correlation between the methods was good (ICC ≥0.79) The time to troughs of 5%, 3% and 1% showed ICCs ≥0.86. For all variables, the most converging limited time point was 6+48 h. Additional time points did not improve the correlation. Despite similar pharmacokinetics, the mean estimated dose for a specific trough level varied from 60% less to 20% more using the population-based approach. The time to 1% and the corresponding dose was sensitive to the baseline assumption. CONCLUSION Our data support the use of population-based PK for patients on simoctocog alfa prophylaxis but also indicates differences, stressing the importance of the sampling scheme and monitoring actual FVIII levels achieved.
Collapse
Affiliation(s)
- Jan Astermark
- Department of Translational Medicine, Lund University, Malmö, Sweden.,Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Malmö, Sweden
| | - Anna Olsson
- Department of Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pierre Chelle
- School of Pharmacy, University of Waterloo, Waterloo, Canada
| | | | | | - Maria Magnusson
- Karolinska Institutet, Coagulation Unit, Department of Hematology, Department of Pediatrics, CLINTEC, Clinical Chemistry & Coagulation, MMK, Karolinska University Hospital, Stockholm, Sweden
| | - Alfonso Iorio
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
27
|
Katragadda S, Neelakantan S, Diao L, Wong N. Population Pharmacokinetic Analysis of Recombinant Factor VIII Fc Fusion Protein in Subjects With Severe Hemophilia A: Expanded to Include Pediatric Subjects. J Clin Pharmacol 2021; 61:889-900. [PMID: 33719084 DOI: 10.1002/jcph.1854] [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: 11/27/2020] [Accepted: 03/04/2021] [Indexed: 01/19/2023]
Abstract
Recombinant factor VIII Fc fusion protein (rFVIIIFc) has been indicated for adults and children with hemophilia A. The objective of this article was to build a population pharmacokinetic (PK) model using adult and pediatric data sets and explore relevant dosing scenarios across all ages. The activity-time profiles of rFVIIIFc from 3 clinical studies (all trials registered at https://www.clinicaltrials.gov: NCT01027377, NCT01181128, and NCT01458106) were characterized, and covariates that determine variability of rFVIIIFc PK in children and adults were identified and implemented. Data sets were pooled to estimate population PK parameters. Simulations were conducted to generate activity-time profiles at steady state (SS). The proportion of subjects maintaining SS trough >1 and >3 IU/dL and time >10 IU/dL were estimated. The rFVIIIFc model was a two-compartment model that identified weight and von Willebrand factor as significant covariates. Model-predicted SS peaks and troughs of rFVIIIFc activity-time profiles confirmed the necessity of modifying dosing in pediatric subjects. The model also predicted that the average subject in the adult and adolescent group dosed with 40 IU/kg every 2 days maintained factor VIII activity >10 IU/dL for the entire duration. Children aged <6 years and aged 6 to <12 years receiving this dose maintained factor VIII activity of >10 IU/dL for nearly two-thirds and three-quarters of their time, respectively. In conclusion, these population PK analyses characterize activity-time profiles for rFVIIIFc among pediatric and adult subjects. The model was used for simulation of clinically relevant dosing scenarios, which can provide better protection and better clinical outcomes.
Collapse
Affiliation(s)
| | | | - Lei Diao
- EpimAb Biotherapeutics, Shanghai, China
| | | |
Collapse
|
28
|
Rurioctocog alfa pegol PK-guided prophylaxis in hemophilia A: results from the phase 3 PROPEL study. Blood 2021; 137:1818-1827. [PMID: 33150384 DOI: 10.1182/blood.2020005673] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/13/2020] [Indexed: 01/19/2023] Open
Abstract
Rurioctocog alfa pegol prophylaxis targeting factor VIII (FVIII) troughs ≥1% has shown to be efficacious with an acceptable safety profile in people with hemophilia A (PwHA). The PROPEL trial compared safety and efficacy of 2 target FVIII troughs in PwHA aged 12 to 65 years, with severe disease, annualized bleeding rate ≥2, and previous FVIII treatment. PwHA were randomized to 12 months' pharmacokinetic (PK)-guided rurioctocog alfa pegol prophylaxis targeting FVIII troughs of 1% to 3% (reference arm) or 8% to 12% (elevated arm); first 6 months was treatment-adjustment period. The primary endpoint was absence of bleeds during the second 6 months, analyzed using multiple imputations (full analysis set [FAS]). In the 1% to 3% and 8% to 12% arms, respectively, point estimates (95% confidence interval) of proportions of PwHA with zero total bleeds were 42% (29% to 55%) and 62% (49% to 75%) in FAS (N = 115; P = .055) and 40% (27% to 55%) and 67% (52% to 81%) in per-protocol analysis set (N = 95; P = .015). Dosing frequency and consumption varied in each arm. Adverse events (AEs) occurred in 70/115 (60.9%) PwHA; serious AEs in 7/115 (6%) PwHA, including 1 treatment-related in 8% to 12% arm (transient anti-FVIII inhibitor). There were no deaths, serious thrombotic events, or AE-related discontinuations. PK-guided prophylaxis was achievable and efficacious in both arms. No new safety signals were observed in the 8% to 12% arm. These results demonstrate elevated FVIII troughs can increase the proportion of PwHA with zero bleeds and emphasize the importance of personalized treatment. This trial was registered at www.clinicaltrials.gov as #NCT02585960.
Collapse
|
29
|
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
|
30
|
Atsou S, Furlan F, Duchemin J, Ellouze S, Sourdeau É, Launois A, Roussel-Robert V, Stieltjes N, Combe S, Fontenay M, Curis E, Jourdi G. Pharmacodynamics of eftrenonacog-alfa (rFIX-Fc) in severe hemophilia B patients: A real-life study. Eur J Pharmacol 2020; 891:173764. [PMID: 33249076 DOI: 10.1016/j.ejphar.2020.173764] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 01/26/2023]
Abstract
Eftrenonacog-alfa is a recombinant factor IX-Fc fusion protein increasingly prescribed in hemophilia B patients. We aimed to assess its pharmacodynamics (PD) in real-life setting via FIX activity measurement and thrombin generation assay (TGA). Sixty samples from 15 severe hemophilia B treated patients were collected at different time points. FIX activity was measured using product-specific one-stage clotting assay (reference method) and two chromogenic assays (CSA) (Biophen FIX and Rox FIX). TGA was triggered with 1 pM tissue factor. Five parameters were analyzed: lag time (LT), time to peak (TTP), peak height (PH), endogenous thrombin potential (ETP), and velocity. PD models were built to characterize their relationships with FIX activity, using mixed effects models. Mean trough FIX level was estimated at 4.64 (±1.50) IU/dl with a recovery at 0.78 (±0.16) IU/dl per 1 IU/kg injected dose. FIX activity ranged between 1 and 86 IU/dl with 21.5 IU/dl median value. Biophen FIX and Rox FIX allowed reliable measurements except in samples with FIX <20 IU/dl in which values were underestimated (delta >30%). PD models revealed that velocity was the most sensitive TGA parameter to FIX activity followed by PH, ETP, TTP and finally LT. Following FIX activity peak after eftrenonacog-alfa injection, velocity decreased first, followed by PH then ETP. Both CSA failed to accurately measure FIX in severe hemophilia B patients receiving eftrenonacog-alfa throughout the measuring range. TGA could be an additional valuable tool to evaluate hemostasis balance in treated patients.
Collapse
Affiliation(s)
- Senade Atsou
- Service D'Hématologie Biologique, AP-HP.Centre-Université de Paris, Hôpital Cochin, F-75014, Paris, France
| | - Fiona Furlan
- Service D'Hématologie Biologique, AP-HP.Centre-Université de Paris, Hôpital Cochin, F-75014, Paris, France
| | - Jérôme Duchemin
- Service D'Hématologie Biologique, AP-HP.Centre-Université de Paris, Hôpital Cochin, F-75014, Paris, France
| | - Syrine Ellouze
- Service D'Hématologie Biologique, AP-HP.Centre-Université de Paris, Hôpital Cochin, F-75014, Paris, France
| | - Élise Sourdeau
- Service D'Hématologie Biologique, AP-HP.Centre-Université de Paris, Hôpital Cochin, F-75014, Paris, France
| | - Amélie Launois
- Service D'Hématologie Biologique, AP-HP.Centre-Université de Paris, Hôpital Cochin, F-75014, Paris, France
| | - Valérie Roussel-Robert
- Centre de Ressources et de Compétences des Maladies Hémorragiques Constitutionnelles, AP-HP.Centre-Université de Paris, Hôpital Cochin, F-75014, Paris, France
| | - Natalie Stieltjes
- Centre de Ressources et de Compétences des Maladies Hémorragiques Constitutionnelles, AP-HP.Centre-Université de Paris, Hôpital Cochin, F-75014, Paris, France
| | - Sophie Combe
- Centre de Ressources et de Compétences des Maladies Hémorragiques Constitutionnelles, AP-HP.Centre-Université de Paris, Hôpital Cochin, F-75014, Paris, France
| | - Michaela Fontenay
- Service D'Hématologie Biologique, AP-HP.Centre-Université de Paris, Hôpital Cochin, F-75014, Paris, France; Université de Paris, Faculté de Santé, F-75006, Paris, France
| | - Emmanuel Curis
- Laboratoire de Biomathématiques, EA 7537, Faculté de Pharmacie, Université de Paris, France; Service de Biostatistiques et Informatique Médicale SBIM, Hôpital Saint Louis, AP-HP, Paris, France
| | - Georges Jourdi
- Service D'Hématologie Biologique, AP-HP.Centre-Université de Paris, Hôpital Cochin, F-75014, Paris, France; Université de Paris, Innovative Therapies in Haemostasis, INSERM UMR-S1140, F-75006, Paris, France.
| |
Collapse
|
31
|
Hermans C, Dolan G. Pharmacokinetics in routine haemophilia clinical practice: rationale and modalities-a practical review. Ther Adv Hematol 2020; 11:2040620720966888. [PMID: 33194165 PMCID: PMC7594230 DOI: 10.1177/2040620720966888] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 09/24/2020] [Indexed: 01/19/2023] Open
Abstract
Prophylactic therapy with exogenous clotting factor concentrates in haemophilia A
and B aims to achieve levels of circulating FVIII or FIX that are adequate for
the prevention or reduction of spontaneous joint bleeding. Historically, a
minimum trough level of at least 1% of the normal levels of circulating clotting
factor has been targeted using standardised protocols. However, clearance of
clotting factor varies between products and patients, and other pharmacokinetic
(PK) parameters such as the frequency and magnitude of peaks may be important
for ensuring optimal coverage. Thus, it is increasingly recognised that an
individualised, PK-based approach to prophylaxis is necessary to achieve optimal
protection. This review focuses on the clinical implications of using PK-guided,
individualised prophylaxis in haemophilia to improve patient outcomes and
considers practical methods of establishing patients’ PK parameters. The most
useful PK parameters will depend on the aim of the specific treatment (e.g.
preventing activity-related and traumatic bleeds or addressing subclinical
bleeding). In clinical practice, lengthy and frequent post-infusion sampling for
PK analysis is costly and a significant burden for patients. However, a Bayesian
analysis allows for the estimation of different PK parameters (e.g. half-life,
factor concentrations over time, etc.) with only a minimum number of samples
(e.g. 4, 24 and 48 h for haemophilia A), by using the patient’s data to adjust a
relevant population PK value towards the actual value. Numerous tools are
available to aid in the practical use of Bayesian PK-guided dosing in the
clinic, including the Web-based Application for the Population Pharmacokinetic
Service hosted by McMaster University, Canada. The PK data can be used to
determine the appropriate prophylaxis regimen for the individual patient, which
can be monitored by assessment of the trough level at each clinic visit. Collection of PK data and subsequent PK-guided dosing should become standard
practice when determining treatment strategies for people with haemophilia.
Collapse
Affiliation(s)
- Cedric Hermans
- Haemostasis and Thrombosis Unit, Division of Adult Haematology, St-Luc University Hospital, Université catholique de Louvain (UCLouvain), Avenue Hippocrate 10, Brussels, 1200, Belgium
| | - Gerry Dolan
- Haemophilia and Thrombosis Centre, St Thomas' Hospital, London, UK
| |
Collapse
|
32
|
Etranacogene dezaparvovec (AMT-061 phase 2b): normal/near normal FIX activity and bleed cessation in hemophilia B. Blood Adv 2020; 3:3241-3247. [PMID: 31698454 DOI: 10.1182/bloodadvances.2019000811] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 09/16/2019] [Indexed: 12/23/2022] Open
Abstract
Etranacogene dezaparvovec (AMT-061) is a recombinant AAV5 vector including a gene cassette containing the factor IX (FIX) Padua variant under the control of a liver-specific promoter. A phase 2b study was conducted to confirm that a single dose of 2 × 1013 genome copies per kilogram of etranacogene dezaparvovec will result in FIX activity ≥5% 6 weeks after dosing. Secondary end points included FIX activity at other time points, bleed frequency, FIX replacement, and safety. Etranacogene dezaparvovec was administered as a single IV infusion to 3 adults with severe to moderately severe hemophilia B. Before treatment, participants had low levels of preexisting neutralizing antibodies to AAV5. This article reports a planned 26-week interim assessment. At week 6, mean FIX activity was 31% (23.9%-37.8%), increasing to 47% (33.2%-57.0%) at 26 weeks, with 2 subjects exhibiting sustained activity >40%. Consistent with the FIX activity, etranacogene dezaparvovec was associated with a complete bleed cessation with no need for FIX replacement therapy up to 26 weeks. Etranacogene dezaparvovec was generally well tolerated. No clinically significant elevations in levels of liver enzymes or inflammatory markers were observed, and no use of corticosteroids related to treatment was required. In individuals with severe to moderately severe hemophilia B, etranacogene dezaparvovec resulted in clinically relevant increases in FIX activity, cessation of bleeds, and abrogation of the need for FIX replacement, despite the presence of preexisting anti-AAV5 neutralizing antibodies detected by using a highly sensitive luciferase assay. Consistency of results in the 3 participants supported an expanded evaluation of the safety/efficacy of etranacogene dezaparvovec in the HOPE-B (Health Outcomes With Padua Gene; Evaluation in Hemophilia-B) phase 3 trial. The current trial was registered at www.clinicaltrials.gov as #NCT03489291.
Collapse
|
33
|
Gelbenegger G, Schoergenhofer C, Knoebl P, Jilma B. Bridging the Missing Link with Emicizumab: A Bispecific Antibody for Treatment of Hemophilia A. Thromb Haemost 2020; 120:1357-1370. [PMID: 32717759 PMCID: PMC7649063 DOI: 10.1055/s-0040-1714279] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Hemophilia A, characterized by absent or ineffective coagulation factor VIII (FVIII), is a serious bleeding disorder that entails severe and potentially life-threatening bleeding events. Current standard therapy still involves replacement of FVIII, but is often complicated by the occurrence of neutralizing alloantibodies (inhibitors). Management of patients with inhibitors is challenging and necessitates immune tolerance induction for inhibitor eradication and the use of bypassing agents (activated prothrombin complex concentrates or recombinant activated factor VII), which are expensive and not always effective. Emicizumab is the first humanized bispecific monoclonal therapeutic antibody designed to replace the hemostatic function of activated FVIII by bridging activated factor IX and factor X (FX) to activate FX and allow the coagulation cascade to continue. In the majority of hemophilic patients with and without inhibitors, emicizumab reduced the annualized bleeding rate to almost zero in several clinical trials and demonstrated a good safety profile. However, the concurrent use of emicizumab and activated prothrombin complex concentrate imposes a high risk of thrombotic microangiopathy and thromboembolic events on patients and should be avoided. Yet, the management of breakthrough bleeds and surgery remains challenging with only limited evidence-based recommendations being available. This review summarizes published clinical trials and preliminary reports of emicizumab and discusses the clinical implications of emicizumab in treatment of hemophilia A.
Collapse
Affiliation(s)
- Georg Gelbenegger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Paul Knoebl
- Division of Hematology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
34
|
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
|
35
|
Fischer K, Ljung R. WITHDRAWN: Primary prophylaxis in haemophilia care: Guideline update 2016. Blood Cells Mol Dis 2020; 80:102172. [PMID: 28363466 DOI: 10.1016/j.bcmd.2017.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/08/2017] [Accepted: 03/13/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Kathelijn Fischer
- Van Creveld kliniek, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rolf Ljung
- Lund University, Dept. of Clinical Sciences Lund, Paediatrics, Lund, Sweden; Skåne University Hospital, Department of Paediatrics and Malmö Centre for Thrombosis and Haemostasis, Malmö, Sweden.
| |
Collapse
|
36
|
Identification of the Profile of the Patients with Hemophilia B Eligible for Treatment with Nonacog Alfa Once-Weekly. REPORTS 2020. [DOI: 10.3390/reports3010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
This study aimed to identify the characteristics of patients with hemophilia B eligible for once-weekly treatment with Nonacog alfa. Methods: A survey was conducted in 14 Hemophilia (HCs) of Italy. These centers were given a questionnaire consisting of ten closed multiple-choice questions. The centers were asked: (a) the percentages of their hemophilia B (HB) patients undergoing replacement therapy, “On-demand”, or weekly prophylaxis, (b) the criteria guiding the monitoring of patients, the advantages according to the age of patients, and (c) the obstacles to prophylaxis. The percentage of patients receiving “On-demand” (OD) treatment or continuous prophylaxis (prophy) differed depending on patient age and the severity of the disease. Only 57% of HCs provided “On-demand” therapy to the mild HB patients, about 93% to moderate ones, of whom 43% on prophylaxis. About 78% of patients <6 years old, were on treatment in 9 out of 14 HCs, by prophylaxis 66.7% and 33.3% by On-demand. In the 6–18 age group, 90.1% of HCs treated HB patients with prophylaxis, 42.8% in the 18–30 age range. On-demand treatment was the therapy of choice in 61.5% of HCs for patients aged 30–65 years. In total, 64% of the HCs assigned the maximum score to bleeding frequency, especially in the <6 and 6–18 age groups. Bleeding severity was also taken into significant consideration, particularly in subjects up to 30 years old. The scores regarding venous access were distributed relatively evenly throughout all age groups. The majority of the centers attributed a medium-high score to treatment compliance, especially in the 6–65 age range. In actuality, 55% of HCs attributed pro-thrombotic comorbidity a low score in the 18–30 age group, whereas 81% gave pro-hemorrhagic comorbidity a high rating in patients aged >65 years old. Many centers assigned a medium-high score to the baseline concentration of FIX level at diagnosis in all age groups. Most HCs attributed a medium-high score to type of genetic mutation in the younger age groups. As for socio-cultural barriers and quality of life, the majority of respondents gave a medium-high score in all age groups. For periodic monitoring of patients receiving continuous prophylaxis, 59% of the centers reported using clinical assessment. With regard to prophylaxis administration method, the majority of hemophiliacs were given infusions twice weekly, while as regards to the dose of FIX concentrate delivered, 50% of the centers reported administering prophylaxis once-weekly at a dose ranging from 5–100 IU/kg in 10–50% of HB patients. Thus, 93% of the centers reported using a dose of 25–50 IU/kg for twice-weekly prophylaxis in 6–100% of the patients. The majority of centers (86%) believe that, in a program of early primary prevention, once-weekly treatment with nonacog alfa may represent an alternative strategy to dose escalation. The results show that patients with mild hemophilia, with functional musculoskeletal status and difficulties with venous access, are candidates for once-weekly prophylaxis with nonacog alfa. For such patients, this regimen can improve treatment compliance and quality of life.
Collapse
|
37
|
Zhou JY, Barnes RFW, Foster G, Iorio A, Cramer TJ, von Drygalski A. Joint Bleeding Tendencies in Adult Patients With Hemophilia: It's Not All Pharmacokinetics. Clin Appl Thromb Hemost 2020; 25:1076029619862052. [PMID: 31298044 PMCID: PMC6714908 DOI: 10.1177/1076029619862052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Hemophilic arthropathy from joint bleeding remains a complication with major morbidity in the increasingly aging patients with hemophilia. Prophylactic clotting factor infusions, based on pharmacokinetic dosing to reduce bleeding rates, are being explored more and more. However, there is little evidence on the benefits of pharmacokinetic dosing in direct association with bleeding events. Here, we prospectively followed a cohort of adult patients with hemophilia A and B (n = 26) and arthropathic joints on various clotting factor products over a period of 2 years with clinical and radiographic joint health assessments, frequent joint ultrasound, and pharmacokinetic studies. Joint bleeds and synovitis with synovial vascularity changes were objectively diagnosed by musculoskeletal ultrasound and power Doppler and analyzed in relation to pharmacokinetic, joint- and patient-specific parameters. Results revealed that, contrary to common beliefs, bleeding episodes were not readily explained by pharmacokinetic features, as they were not associated with more time spent below certain clotting factor thresholds. Joint bleeding was found to be associated with prominent vascularity changes, suggesting that vascular remodeling and leakiness may contribute to joint bleeding that cannot be prevented by clotting factor replacement alone.
Collapse
Affiliation(s)
- Jenny Y Zhou
- 1 Division of Hematology/Oncology, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Richard F W Barnes
- 1 Division of Hematology/Oncology, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Gary Foster
- 2 Department of Health Evidence, Research Methodology and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Alfonso Iorio
- 2 Department of Health Evidence, Research Methodology and Impact, McMaster University, Hamilton, Ontario, Canada.,3 Department of Medicine, McMaster-Bayer Endowed Research Chair in Clinical Epidemiology of Congenital Bleeding Disorders, McMaster University, Hamilton, Ontario, Canada
| | - Thomas J Cramer
- 1 Division of Hematology/Oncology, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Annette von Drygalski
- 1 Division of Hematology/Oncology, Department of Medicine, University of California, San Diego, San Diego, CA, USA.,4 Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
38
|
Chowdary P. Extended half-life recombinant products in haemophilia clinical practice - Expectations, opportunities and challenges. Thromb Res 2019; 196:609-617. [PMID: 31883700 DOI: 10.1016/j.thromres.2019.12.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/03/2019] [Accepted: 12/18/2019] [Indexed: 02/06/2023]
Abstract
Extended half-life (EHL) products have shown robust efficacy in clinical trials, whilst allowing for less intense treatment regimens when compared with standard half-life products. Regimen optimisation with EHL products could lead to further improvements in bleeding rates, quality of life and reductions in treatment burden. Patients now expect good efficacy, a lower treatment burden and equivalent safety when compared with standard half-life products. As our knowledge base grows these expectations have evolved and targeting an annualised bleeding rate of zero has become a more realistic clinical goal. Personalised prophylaxis can help patients achieve these goals. However, a number of challenges still remain, including cost, challenges in predicting outcomes for patients and differences in patients' and clinicians' expectations. When switching a patient, comprehensive patient care can reduce the impact of these issues. This review presents in brief the protein therapeutics with an extended half-life, including key trial results, challenges of chronic care that impact on patients' outcomes and how the modified proteins might help address some of these issues. In addition, practical steps for managing the switch to EHL products are presented.
Collapse
Affiliation(s)
- Pratima Chowdary
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free Hospital, London, UK.
| |
Collapse
|
39
|
McEneny-King A, Yeung CH, Edginton AN, Iorio A, Croteau SE. Clinical application of Web Accessible Population Pharmacokinetic Service-Hemophilia (WAPPS-Hemo): Patterns of blood sampling and patient characteristics among clinician users. Haemophilia 2019; 26:56-63. [PMID: 31742831 DOI: 10.1111/hae.13882] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Use of population pharmacokinetics (PopPK) to facilitate PK-informed prophylaxis in clinical practice has gained momentum among haemophilia providers due to the accessibility of tools such as the Web Accessible Population Pharmacokinetic Service-Hemophilia (WAPPS-Hemo) and availability of extended half-life (EHL) factor concentrates. It is unknown how clinicians implement PopPK. AIM To investigate the evolution of PopPK use in clinical practice by comparing blood sampling strategies, patient features, and factor group between initial and recent periods of WAPPS-Hemo availability. METHODS PK data for haemophilia A and haemophilia B patients from two time periods were extracted from the WAPPS-Hemo database: early availability (10/2015-09/2016) and recent use (10/2017-09/2018). We compared patient characteristics (age, body weight, haemophilia type), product type and dose, and blood sampling times between the time frames. RESULTS Over 1900 eligible infusions were submitted to WAPPS-Hemo during the periods studied, with 85% representing FVIII concentrates. In the recent cohort, PK profiles were requested for younger patients (median age 18 vs 26 years), with increased proportional EHL FVIII use (29% vs 14% of infusions). High-use centres generally submitted fewer blood samples per infusion than non-high-use centres, although the number of samples collected by non-high-use centres decreased significantly over time. During both periods, blood sample timing was generally consistent with ISTH recommended windows. CONCLUSION The use of WAPPS-Hemo by haemophilia providers grew by over threefold between the time periods investigated. While sampling times have included key time points proposed first by Björkman since early WAPPS-Hemo usage, a trend towards minimizing sampling was observed.
Collapse
Affiliation(s)
| | - Cindy Ht Yeung
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | | | - Alfonso Iorio
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada.,Department of Medicine, Clinical Epidemiology of Congenital Bleeding Disorders, McMaster University, Hamilton, ON, Canada
| | - Stacy E Croteau
- Boston Hemophilia Center, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
40
|
Spronck EA, Liu YP, Lubelski J, Ehlert E, Gielen S, Montenegro-Miranda P, de Haan M, Nijmeijer B, Ferreira V, Petry H, van Deventer SJ. Enhanced Factor IX Activity following Administration of AAV5-R338L "Padua" Factor IX versus AAV5 WT Human Factor IX in NHPs. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:221-231. [PMID: 31709273 PMCID: PMC6834974 DOI: 10.1016/j.omtm.2019.09.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/19/2019] [Indexed: 10/29/2022]
Abstract
Gene therapy for severe hemophilia B is advancing and offers sustained disease amelioration with a single treatment. We have reported the efficacy and safety of AMT-060, an investigational gene therapy comprising an adeno-associated virus serotype 5 capsid encapsidating the codon-optimized wild-type human factor IX (WT hFIX) gene with a liver-specific promoter, in patients with severe hemophilia B. Treatment with 2 × 1013 gc/kg AMT-060 showed sustained and durable FIX activity of 3%-13% and a substantial reduction in spontaneous bleeds without T cell-mediated hepatoxicity. To achieve higher FIX activity, we modified AMT-060 to encode the R338L "Padua" FIX variant that has increased specific activity (AMT-061). We report the safety and increased FIX activity of AMT-061 in non-human primates. Animals (n = 3/group) received intravenous AMT-060 (5 × 1012 gc/kg), AMT-061 (ranging from 5 × 1011 to 9 × 1013 gc/kg), or vehicle. Human FIX protein expression, FIX activity, and coagulation markers including D-dimer and thrombin-antithrombin complexes were measured. At equal doses, AMT-060 and AMT-061 resulted in similar human FIX protein expression, but FIX activity was 6.5-fold enhanced using AMT-061. Both vectors show similar safety and transduction profiles. Thus, AMT-061 holds great promise as a more potent FIX replacement gene therapy with a favorable safety profile.
Collapse
Affiliation(s)
- Elisabeth A Spronck
- uniQure biopharma B.V., Paasheuvelweg 25A, 1105 BP Amsterdam, the Netherlands
| | - Ying Poi Liu
- uniQure biopharma B.V., Paasheuvelweg 25A, 1105 BP Amsterdam, the Netherlands
| | - Jacek Lubelski
- uniQure biopharma B.V., Paasheuvelweg 25A, 1105 BP Amsterdam, the Netherlands
| | - Erich Ehlert
- uniQure biopharma B.V., Paasheuvelweg 25A, 1105 BP Amsterdam, the Netherlands
| | - Sander Gielen
- uniQure biopharma B.V., Paasheuvelweg 25A, 1105 BP Amsterdam, the Netherlands
| | | | - Martin de Haan
- uniQure biopharma B.V., Paasheuvelweg 25A, 1105 BP Amsterdam, the Netherlands
| | - Bart Nijmeijer
- uniQure biopharma B.V., Paasheuvelweg 25A, 1105 BP Amsterdam, the Netherlands
| | - Valerie Ferreira
- uniQure biopharma B.V., Paasheuvelweg 25A, 1105 BP Amsterdam, the Netherlands
| | - Harald Petry
- uniQure biopharma B.V., Paasheuvelweg 25A, 1105 BP Amsterdam, the Netherlands
| | - Sander J van Deventer
- uniQure biopharma B.V., Paasheuvelweg 25A, 1105 BP Amsterdam, the Netherlands.,Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| |
Collapse
|
41
|
Balen EC, Krawczyk M, Gue D, Jackson S, Gouw SC, Bom JG, Sawatzky R. Patient‐centred care in haemophilia: Patient perspectives on visualization and participation in decision‐making. Haemophilia 2019; 25:938-945. [DOI: 10.1111/hae.13830] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 06/25/2019] [Accepted: 07/09/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Erna C. Balen
- Department of Clinical Epidemiology Leiden University Medical Center Leiden The Netherlands
| | - Marian Krawczyk
- Lord Kelvin Adam Smith Fellow, School of Interdisciplinary Studies University of Glasgow Glasgow UK
- School of Nursing Trinity Western University Langley Canada
- Centre for Health Evaluation & Outcome Sciences Providence Health Care Research Institute Vancouver Canada
| | - Deborah Gue
- British Columbia Provincial Bleeding Disorders Program ‐ Adult Division St. Paul’s Hospital Vancouver Canada
| | - Shannon Jackson
- British Columbia Provincial Bleeding Disorders Program ‐ Adult Division St. Paul’s Hospital Vancouver Canada
| | - Samantha C. Gouw
- Department of Clinical Epidemiology Leiden University Medical Center Leiden The Netherlands
- Department of Pediatric Hematology, Emma Children’s Hospital Amsterdam UMC, University of Amsterdam Amsterdam The Netherlands
| | - Johanna G. Bom
- Department of Clinical Epidemiology Leiden University Medical Center Leiden The Netherlands
- Sanquin Research Center for Clinical Transfusion Research Leiden The Netherlands
| | - Richard Sawatzky
- School of Nursing Trinity Western University Langley Canada
- Centre for Health Evaluation & Outcome Sciences Providence Health Care Research Institute Vancouver Canada
- Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| |
Collapse
|
42
|
Abrantes JA, Solms A, Garmann D, Nielsen EI, Jönsson S, Karlsson MO. Relationship between factor VIII activity, bleeds and individual characteristics in severe hemophilia A patients. Haematologica 2019; 105:1443-1453. [PMID: 31371418 PMCID: PMC7193498 DOI: 10.3324/haematol.2019.217133] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 07/23/2019] [Indexed: 01/19/2023] Open
Abstract
Pharmacokinetic-based prophylaxis of replacement factor VIII (FVIII) products has been encouraged in recent years, but the relationship between exposure (factor VIII activity) and response (bleeding frequency) remains unclear. The aim of this study was to characterize the relationship between FVIII dose, plasma FVIII activity, and bleeding patterns and individual characteristics in severe hemophilia A patients. Pooled pharmacokinetic and bleeding data during prophylactic treatment with BAY 81-8973 (octocog alfa) were obtained from the three LEOPOLD trials. The population pharmacokinetics of FVIII activity and longitudinal bleeding frequency, as well as bleeding severity, were described using non-linear mixed effects modeling in NONMEM. In total, 183 patients [median age 22 years (range, 1-61); weight 60 kg (11-124)] contributed with 1,535 plasma FVIII activity observations, 633 bleeds and 11 patient/study characteristics [median observation period 12 months (3.1-13.1)]. A parametric repeated time-to-categorical bleed model, guided by plasma FVIII activity from a 2-compartment population pharmacokinetic model, described the time to the occurrence of bleeds and their severity. Bleeding probability decreased with time of study, and a bleed was not found to affect the time of the next bleed. Several covariate effects were identified, including the bleeding history in the 12-month pre-study period increasing the bleeding hazard. However, unexplained inter-patient variability in the phenotypic bleeding pattern remained large (111%CV). Further studies to translate the model into a tool for dose individualization that considers the individual bleeding risk are required. Research was based on a post-hoc analysis of the LEOPOLD studies registered at clinicaltrials.gov identifiers: 01029340, 01233258 and 01311648.
Collapse
Affiliation(s)
- João A Abrantes
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | - Elisabet I Nielsen
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Siv Jönsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Mats O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
43
|
Maji D, Nayak L, Martin J, Sekhon UDS, Sen Gupta A, Mohseni P, Suster MA, Ahuja SP. A novel, point-of-care, whole-blood assay utilizing dielectric spectroscopy is sensitive to coagulation factor replacement therapy in haemophilia A patients. Haemophilia 2019; 25:885-892. [PMID: 31282024 DOI: 10.1111/hae.13799] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/06/2019] [Accepted: 05/14/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Reliable monitoring of coagulation factor replacement therapy in patients with severe haemophilia, especially those with inhibitors, is an unmet clinical need. While useful, global assays, eg thromboelastography (TEG), rotational thromboelastometry (ROTEM) and thrombin generation assay (TGA), are cumbersome to use and not widely available. OBJECTIVE To assess the utility of a novel, point-of-care, dielectric microsensor - ClotChip - to monitor coagulation factor replacement therapy in patients with haemophilia A, with and without inhibitors. METHODS The ClotChip Tpeak parameter was assessed using whole-blood samples from children with severe haemophilia A, with (n = 6) and without (n = 12) inhibitors, collected pre- and postcoagulation factor replacement therapy. ROTEM, TGA and chromogenic FVIII assays were also performed. Healthy children (n = 50) served as controls. RESULTS ClotChip Tpeak values exhibited a significant decrease for samples collected postcoagulation factor replacement therapy as compared to baseline (pretherapy) samples in patients with and without inhibitors. A difference in Tpeak values was also noted at baseline among severe haemophilia A patients with inhibitors as compared to those without inhibitors. ClotChip Tpeak parameter exhibited a very strong correlation with clotting time (CT) of ROTEM, endogenous thrombin potential (ETP) and peak thrombin of TGA, and FVIII clotting activity. CONCLUSIONS ClotChip is sensitive to coagulation factor replacement therapy in patients with severe haemophilia A, with and without inhibitors. ClotChip Tpeak values correlate very well with ROTEM, TGA and FVIII assays, opening up possibilities for its use in personalized coagulation factor replacement therapy in haemophilia.
Collapse
Affiliation(s)
- Debnath Maji
- Department of Electrical Engineering and Computer Science, Case Western Reserve University, Cleveland, Ohio
| | - Lalitha Nayak
- Division of Hematology/Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Janet Martin
- Division of Pediatric Hematology/Oncology, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, Ohio
| | - Ujjal D S Sekhon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Pedram Mohseni
- Department of Electrical Engineering and Computer Science, Case Western Reserve University, Cleveland, Ohio
| | - Michael A Suster
- Department of Electrical Engineering and Computer Science, Case Western Reserve University, Cleveland, Ohio
| | - Sanjay P Ahuja
- Division of Pediatric Hematology/Oncology, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
44
|
Carcao M, Escuriola‐Ettingshausen C, Santagostino E, Oldenburg J, Liesner R, Nolan B, Bátorová A, Haya S, Young G. The changing face of immune tolerance induction in haemophilia A with the advent of emicizumab. Haemophilia 2019; 25:676-684. [PMID: 31033112 PMCID: PMC6850066 DOI: 10.1111/hae.13762] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 03/20/2019] [Accepted: 03/28/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION As a result of the new treatment paradigm that the haemophilia community will face with the availability of novel (non-factor) therapies, an updated consensus on ITI recommendations and inhibitor management strategies is needed. AIM The Future of Immunotolerance Treatment (FIT) group was established to contemplate, determine and recommend the best management options for patients with haemophilia A and inhibitors. DISCUSSION AND CONCLUSIONS Despite the considerable success of emicizumab in the management of inhibitor patients, the FIT group still sees the importance of eradicating inhibitors. However, the availability of emicizumab and other non-factor therapies in the future might impact greatly on how ITI is undertaken. Theoretically, concomitant use of emicizumab and FVIII might allow emicizumab to effectively prevent bleeding with lower dose ITI regimens. This might allow for the greater adoption of low-dose/low-frequency FVIII ITI regimens, which may result in a reduced need for central venous access devices while still maintaining a reasonable likelihood of ITI success. The FIT group proposes a new management algorithm for current ITI (without emicizumab) and a hypothetical new approach with the availability of emicizumab. As there are no published data regarding the concomitant use of emicizumab and FVIII for ITI, the FIT Expert group encourages the undertaking of properly conducted prospective studies to explore these approaches further.
Collapse
Affiliation(s)
- Manuel Carcao
- Division of Haematology/Oncology, Department of Paediatrics and Child Health Evaluative Sciences, Research Institute, The Hospital for Sick ChildrenUniversity of TorontoTorontoOntarioCanada
| | | | - Elena Santagostino
- Maggiore Hospital PoliclinicoAngelo Bianchi Bonomi Hemophilia and Thrombosis CenterMilanItaly
| | - Johannes Oldenburg
- Institute of Experimental Haematology and Transfusion MedicineUniversity of BonnBonnGermany
| | - Ri Liesner
- Haemophilia Centre, Great Ormond Street Hospital for ChildrenNHS Trust Haemophilia CentreLondonUK
| | | | - Angelika Bátorová
- Department of Haematology and Transfusion Medicine and National Haemophilia CentreUniversity Hospital, Comenius UniversityBratislavaSlovakia
| | - Saturnino Haya
- Unit for Congenital Bleeding DisordersHospital Universitario y Politécnico La FeValenciaSpain
| | - Guy Young
- Children's Hospital Los AngelesUniversity of Southern California Keck School of MedicineLos AngelesCalifornia
| | | |
Collapse
|
45
|
Ar MC, Balkan C, Kavaklı K. Extended Half-Life Coagulation Factors: A New Era in the Management of Hemophilia Patients. Turk J Haematol 2019; 36:141-154. [PMID: 31088040 PMCID: PMC6682782 DOI: 10.4274/tjh.galenos.2019.2018.0393] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Despite effective factor replacement and various treatment schedules, there remain several challenges and unmet needs in the prophylactic treatment of hemophilia limiting its adoption and thereby posing an increased risk of spontaneous bleeding. In this regard, extended half-life (EHL) recombinant factor VIII (rFVIII) and factor IX (rFIX) products promise optimal prophylaxis by decreasing the dose frequency, increasing the compliance, and improving the quality of life without compromising safety and efficacy. EHL products might lead to higher trough levels without increasing infusion frequency, or could facilitate the ability to maintain trough levels while reducing infusion frequency. This paper aims to provide a comprehensive review of the rationale for developing EHL coagulation factors and their utility in the management of hemophilia, with special emphasis on optimal techniques for half-life extension and criteria for defining EHL coagulation factors, as well as indications, efficacy, and safety issues of the currently available EHL-rFVIII and EHL-rFIX products. Potential impacts of these factors on quality of life, health economics, and immune tolerance treatment will also be discussed alongside the challenges in pharmacokinetic-driven prophylaxis and difficulties in monitoring the EHL products with laboratory assays.
Collapse
Affiliation(s)
- Muhlis Cem Ar
- İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine Department of Internal Medicine, Division of Hematology, İstanbul, Turkey
| | - Can Balkan
- Ege University Faculty of Medicine, Department of Pediatrics, Division of Hemato-Oncology, İzmir, Turkey
| | - Kaan Kavaklı
- Ege University Faculty of Medicine, Department of Pediatrics, Division of Hemato-Oncology, İzmir, Turkey
| |
Collapse
|
46
|
Escuriola Ettingshausen C, Hegemann I, Simpson ML, Cuker A, Kulkarni R, Pruthi RK, Garly M, Meldgaard RM, Persson P, Klamroth R. Favorable pharmacokinetics in hemophilia B for nonacog beta pegol versus recombinant factor IX-Fc fusion protein: A randomized trial. Res Pract Thromb Haemost 2019; 3:268-276. [PMID: 31011711 PMCID: PMC6462750 DOI: 10.1002/rth2.12192] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/09/2019] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Nonacog beta pegol (N9-GP) and recombinant factor IX-Fc fusion protein (rFIXFc) are extended half-life rFIX compounds. We report the first single-dose pharmacokinetic trial of N9-GP and rFIXFc. PATIENTS/METHODS Paradigm 7 was a multicenter, open-label, randomized, crossover trial in previously treated (>150 exposure days) adults with congenital hemophilia B (FIX activity ≤2%). Patients received single intravenous injections (50 IU/kg) of N9-GP and rFIXFc with at least 21 days between doses. Plasma FIX activity, predose, and at serial time points up to 240 hours postdose, was measured using validated one-stage clotting assays (SynthAFax for N9-GP; Actin FSL for rFIXFc) and a chromogenic assay (ROX factor IX) with normal human plasma as calibrator. The primary endpoint was area under the FIX activity-time curve from 0 to infinity, dose-normalized to 50 IU/kg (AUC0-inf,norm). RESULTS Fifteen patients received study treatment. Based on FIX activity results from the one-stage clotting assays, estimated AUC0-inf,norm was significantly greater for N9-GP than rFIXFc (ratio: 4.39; P < 0.0001, based on a two-sided test on 5% significance level). In addition, N9-GP had a longer terminal half-life, two times higher incremental recovery at 30 minutes and maximum FIX activity (dose-normalized to 50 IU/kg) and six times higher FIX activity at 168 hours than rFIXFc. These findings were largely comparable with the chromogenic assay data and are consistent with published data for each compound. CONCLUSIONS In this comparison, N9-GP demonstrated favorable pharmacokinetic characteristics versus rFIXFc, helping clinicians to understand differences between N9-GP and rFIXFc. REGISTRATION This trial is registered with clinicaltrials.gov (NCT03075670) and the European Clinical Trials Database (EudraCT: 2016-001149-25).
Collapse
Affiliation(s)
| | - Inga Hegemann
- Division of HaematologyZürich University HospitalZürichSwitzerland
| | - Mindy L. Simpson
- Pediatric Hematology/OncologyRush University Medical CenterChicagoILUSA
| | - Adam Cuker
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Roshni Kulkarni
- Department of Pediatrics and Human DevelopmentMichigan State UniversityEast LansingMIUSA
| | | | | | | | | | - Robert Klamroth
- Department for Internal MedicineVascular Medicine and HaemostaseologyVivantes KlinikumBerlinGermany
| |
Collapse
|
47
|
Megías-Vericat J, Bonanad S, Haya S, Cid A, Marqués M, Monte E, Pérez-Alenda S, Bosch P, Querol F, Poveda J. Bayesian pharmacokinetic-guided prophylaxis with recombinant factor VIII in severe or moderate haemophilia A. Thromb Res 2019; 174:151-162. [DOI: 10.1016/j.thromres.2018.12.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/17/2018] [Accepted: 12/31/2018] [Indexed: 01/19/2023]
|
48
|
Versloot O, Berntorp E, Petrini P, Ljung R, Astermark J, Holmström M, de Kleijn P, Fischer K. Sports participation and physical activity in adult Dutch and Swedish patients with severe haemophilia: A comparison between intermediate- and high-dose prophylaxis. Haemophilia 2019; 25:244-251. [PMID: 30690833 PMCID: PMC6850651 DOI: 10.1111/hae.13683] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/14/2018] [Accepted: 01/02/2019] [Indexed: 12/27/2022]
Abstract
Introduction Differences in treatment and outcome have been reported for persons with haemophilia (PWH) on intermediate‐dose (Dutch) and high‐dose (Swedish) prophylaxis, but the potential influence of sports participation has not been considered. Aim To compare sports participation and clinical outcome between adult Dutch and Swedish PWH. Methods Self‐reported sports participation (type and frequency per week), physical functioning (SF‐36PF: 100‐0), joint status (HJHS: 0‐144), perceived limitations (HALsum: 100‐0) and physical activity (IPAQ) were recorded. Sports were classified according to National Haemophilia Foundation classification (5 categories, highest two were classified as high‐risk sports). Sports participation and clinical outcome were compared according to country and age (18‐22, 23‐29, 30‐40 years) using non‐parametric tests and Spearman correlations (rho). Results Seventy‐one adult PWH (NL: 43, SWE: 28) completed sports questionnaires (mean age: 26 years). All participants engaged in sports, including 59.2% in high‐risk sports (33.9% twice weekly). Dutch PWH showed a significant age‐related decline in (high‐risk) sports participation (7x/wk in PWH 18‐22 years to 2x/wk in PWH 30‐40 years, P < 0.05), joint health (HJHS: median 2‐15.5, P < 0.01) and physical functioning (SF‐36PF: median 100 to 77.5, P < 0.01), while Swedish did not. Sports participation was not associated with bleeding (Spearman's rho = −0.119). Conclusion All participants reported sports participation, including 59.2% in high‐risk sports. Dutch PWH treated with intermediate‐dose prophylaxis showed an age‐related decline in sports participation, joint status and physical functioning, whereas Swedish PWH on high‐dose prophylaxis did not. Sports participation was not associated with bleeding.
Collapse
Affiliation(s)
- Olav Versloot
- Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Erik Berntorp
- Clinical Coagulation Research Unit, Lund University, Skåne University Hospital, Malmö, Sweden.,Department of Translational Sciences, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Pia Petrini
- Department of Pediatrics, Clinic of Coagulation Disorders, Karolinska Hospital, Stockholm, Sweden
| | - Rolf Ljung
- Clinical Coagulation Research Unit, Lund University and Malmö Centre for Thrombosis and Haemostasis, Skåne University Hospital, Malmö, Sweden
| | - Jan Astermark
- Centre for Thrombosis and Haemostasis, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Margareta Holmström
- Department of Translational Sciences, Faculty of Medicine, Lund University, Malmö, Sweden.,Coagulation Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Piet de Kleijn
- Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Kathelijn Fischer
- Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
49
|
Taylor A, Vendramin C, Oosterholt S, Della Pasqua O, Scully M. Pharmacokinetics of plasma infusion in congenital thrombotic thrombocytopenic purpura. J Thromb Haemost 2019; 17:88-98. [PMID: 30475428 DOI: 10.1111/jth.14345] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Indexed: 11/29/2022]
Abstract
Essentials Congenital thrombotic thrombocytopenic purpura (TTP) is primarily treated with plasma infusion. We present a pharmacokinetic analysis of ADAMTS-13 in six patients following plasma infusion. A median half-life of 130 h was demonstrated, ranging between 82.6 and 189.5 h. Investigation of interindividual clearance of ADAMTS-13 is necessary to optimize treatment. SUMMARY: Background Congenital thrombotic thrombocytopenic purpura (TTP) is defined by persistent severe deficiency of ADAMTS-13 in the absence of anti-ADAMTS-13 inhibitory antibodies, confirmed by mutational analysis. Replacement of the missing protease prevents disease relapse, primarily using plasma infusion (PI). Objectives, patients and methods There is scant evidence regarding optimal dose and frequency of treatment, which tends to be empirically guided. We present a pharmacokinetic analysis of ADAMTS-13 in six patients with congenital TTP on established regimes following PI. Results We found a median clearance of 25.41 mL h-1 and half-life of 130 h, ranging between 82.6 and 189.5 h (3.4-7.9 days, respectively). All patients reached baseline ADAMTS-13 level within 7-10 days post-plasma. Median ADAMTS-13 activity peak post-PI was 24.05 IU dL-1 . Variation was related to elimination rate, which, in turn, was affected by weight and metabolism, but not to von Willebrand factor antigen or activity levels. Using the pharmacokinetic parameters, we simulated individualized protocols based on PI dose or frequency to target hypothetical optimal plasma levels of ADAMTS-13 of 10 and 50 IU dL-1 , respectively. Results suggest a target trough ADAMTS-13 of 10 IU dL-1 is feasible but 50 IU dL-1 would not be achievable taking into account volume required. Conclusions Further work is needed to compare treatment of congenital TTP with PI vs. recombinant ADAMTS-13. PI may provide longer duration of ADAMTS-13 effect, but is limited by plasma volume required, whereas recombinant therapy can provide a higher ADAMTS-13 peak. We propose that investigation of interindividual clearance of ADAMTS-13 is necessary to optimize treatment and provide the rationale for dose and frequency of prophylaxis.
Collapse
Affiliation(s)
- A Taylor
- Haemostasis Research Unit, University College London, London, UK
| | - C Vendramin
- Haemostasis Research Unit, University College London, London, UK
| | - S Oosterholt
- Clinical Pharmacology and Therapeutics Group, University College London, London, UK
| | - O Della Pasqua
- Clinical Pharmacology and Therapeutics Group, University College London, London, UK
| | - M Scully
- Department of Haematology, UCLH and Cardiometabolic Programme-NIHR UCLH/UC BRC London, London, UK
| |
Collapse
|
50
|
Nagao A, Yeung CH, Germini F, Suzuki T. Clinical outcomes in hemophilia A patients undergoing tailoring of prophylaxis based on population-based pharmacokinetic dosing. Thromb Res 2019; 173:79-84. [DOI: 10.1016/j.thromres.2018.11.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/09/2018] [Accepted: 11/15/2018] [Indexed: 01/19/2023]
|