Abstract
Prophylactic factor VIII (FVIII) has dramatically improved haemophilia A treatment, preventing joint bleeding and halting the deterioration of joint status. FVIII products with an extended plasma half-life further improve patients’ quality of life and increase therapeutic adherence. New licensed classes of non-replacement products include prophylactic emicizumab, which is administered subcutaneously up to every 4 weeks. However, this drug is not suitable for acute bleeding episodes or management of major surgery, and long-term data on the impact of emicizumab on joint health, FVIII inhibitor development and thrombotic risk are awaited. Prophylaxis with FVIII replacement remains the standard of care in haemophilia A, with the aim of achieving a level of haemostasis control that allows patients to meet their lifestyle goals.
Keywords: factor VIII, haemophilia A, prophylaxis, standard of care, half-life
Introduction
The availability of factor replacement products has dramatically improved care for people with haemophilia. Where these treatments are accessible and affordable, individuals have a life expectancy similar to that of males in the general population1. Replacement of factor VIII (FVIII) in haemophilia A has been used for decades to control bleeding, including during and after surgery, and for immune tolerance induction (ITI) in patients who develop alloantibodies that neutralise FVIII coagulant activity (inhibitors). The development of inhibitors is the most serious and expensive treatment-related complication2, affecting at least one-third of individuals with severe haemophilia A3. Bypassing agents, such as activated prothrombin complex concentrates (aPCCs) and recombinant activated factor VII (rFVIIa), are generally used to treat acute bleeding in individuals with inhibitors4; however, some evidence supports the use of prophylaxis with bypassing agents in these patients4. ITI with FVIII is currently the only proven method to eradicate FVIII inhibitors, and is successful in approximately 70% of individuals with haemophilia A5–8.
Randomised clinical trials and real-world evidence have demonstrated that prophylaxis prevents joint bleeding and deterioration of joint status9,10, and primary prophylaxis with FVIII has, therefore, been recognised as the standard of care for individuals with severe haemophilia A in countries with adequate resources11. The landmark Joint Outcomes Study by Manco-Johnson et al. provided evidence of an approximately 6-fold reduction in the risk of joint damage in children with severe haemophilia A assigned to prophylaxis compared with those randomly assigned to on-demand treatment10. Prophylactic therapy also reduces the incidence of central nervous system bleeds (intracranial and spinal haematoma), which are less common than joint bleeding but much more life-threatening12,13. Additional advantages of prophylaxis vs on-demand treatment include reduced hospitalisations and absenteeism from school or work, greater participation in social activities and, on the whole, an improved health-related quality of life14–16.
Currently, targeted trough plasma levels of approximately 1–2% FVIII activity are advisable and achievable in the context of prophylaxis programmes in order to prevent breakthrough bleeding, including joint bleeding. However, prophylaxis with higher troughs of approximately 12% may provide better outcomes and avoid disabilities17. The ability to measure FVIII levels in plasma facilitates tailoring of prophylaxis according to patient-agreed lifestyle priorities. Indeed, application of pharmacokinetic measures plays a key role in tailoring management to individual patients18.
With its increasing availability, prophylaxis has evolved, and now focusses on improving a person’s quality of life by reducing the burden of infusions and increasing treatment adherence19. Intravenous administration has been cited as a key barrier to prophylaxis. Drawbacks include the frequent need for a central venous access device and related complications, plus the fear of needles and the related pain9,20,21. Regimens requiring frequent intravenous injections may be a reason for poor adherence to prophylaxis21,22, because standard half-life (SHL) coagulation FVIII products typically involve the burden of 2–4 injections per week.
With these limitations in mind, new treatments with extended plasma half-lives (EHLs) of the infused FVIII were developed to promote more convenient and individualised dosing schedules, with longer intervals between doses or higher threshold levels with the same dosing interval22–26. Several techniques have been used to extend the plasma half-life of currently licensed products. The two main approaches are Fc-fusion technology and PEGylation, which is the conjugation of the FVIII protein with polyethylene glycol (PEG). Here we critically review the advantages of EHL FVIII products and outline their limitations, and discuss the development and licensing of new non-replacement treatment products, such as emicizumab.
Fc-fusion extended plasma half-life product
The recombinant FVIII Fc-fusion protein (rFVIII-Fc; Eloctate®, Sanofi Genzyme, Cambridge, MA, USA), composed of human FVIII fused with a monomeric human immunoglobulin (IgG1) Fc domain24, was the first EHL product to be licensed (2014), with indications for the treatment and prevention of bleeding and for perioperative management of individuals with haemophilia A27,28. In a phase I/IIa safety and pharmacokinetic study in previously treated patients with severe haemophilia A, rFVIII-Fc demonstrated a 1.5–1.7-fold longer plasma half-life compared with a SHL rFVIII24. Results from the phase III studies A-LONG and Kids A-LONG, and interim results from the ongoing extension study ASPIRE, established the long-term efficacy and safety of rFVIII-Fc for the treatment and prevention of bleeding in previously treated patients with severe haemophilia A29–31. The median annualised bleeding rates (ABR) observed for individualised prophylaxis, weekly prophylaxis and episodic treatment arms were 1.6, 3.6 and 33.6, with no bleeding episodes occurring in 45.3% and 17.4% of patients on prophylaxis in arms 1 and 2 (Table I). In Kids A-LONG, participants received twice-weekly rFVIII-Fc (25 IU/kg and 50 IU/kg on days 1 and 4). Median ABR was 1.96 overall30. This low ABR was maintained in ASPIRE, which enroled participants from the A-LONG and Kids A-LONG, with no bleeding events occurring in 22.2–59.3% of patients across the different regimens (Table I)31. rFVIII-Fc was well tolerated and efficacious for the management of perioperative haemostasis across a wide spectrum of major and minor surgery in patients with severe haemophilia enroled on the primary and ASPIRE studies32. During most major surgeries (95.7%), haemostasis was maintained with one rFVIII-Fc infusion. No FVIII inhibitors were observed29–32, although at-risk patients with a history or evidence of FVIII inhibitors, as well as previously untreated patients, were excluded from the pivotal clinical trials. rFVIII-Fc has also been used for ITI in patients with severe haemophilia A and high-titre inhibitors, with varying levels of success33.
Table I.
Product and clinical trial | Regimen | Overall ABR (median) | Zero bleeding ratesa |
---|---|---|---|
rFVIII-Fc (Eloctate®) | |||
| |||
A-LONG29 | Individualised prophylaxis (25–65 IU/kg every 3–5 days) (n=117) | 1.6 | 45.3% |
| |||
QW prophylaxis (65 IU/kg) (n=23) | 3.6 | 17.4% | |
| |||
Episodic treatment (10–50 IU/kg) (n=23) | 33.6 | 0 | |
| |||
Kids A-LONG30 | Twice-weekly prophylaxis (25 IU/kg and 50 IU/kg on days 1 and 4) (n=69) | 1.96 | NR |
| |||
ASPIRE (extension study)b,31 | A-LONG patients: | ||
Individualised prophylaxis (n=108) | 0.66 | 38.9% | |
| |||
QW prophylaxis (n=27) | 2.03 | 22.2% | |
| |||
Modified prophylaxis (n=17) | 1.97 | 23.5% | |
| |||
Episodic treatment (n=14) | 18.36 | - | |
Kids A-LONG patients: | |||
Individualised prophylaxis <6 years (n=29) | 0 | NR | |
| |||
Individualised prophylaxis 6–12 years (n=30) | 1.54 | NR | |
| |||
Individualised prophylaxis <6–12 years (n=59) | NR | 59.3% | |
| |||
Modified prophylaxis <6 years (n=1) | 6.55 | NR | |
| |||
Modified prophylaxis 6–12 years (n=1) | 0 | NR | |
| |||
Modified prophylaxis <6–12 years (n=2) | NR | 50% | |
| |||
BAX 855 (Adynovate®) | |||
| |||
PROLONG-ATE23 | Twice-weekly prophylaxis (45±5 IU/kg for 6 months [≥50 EDs]) (n=120) | 1.9 | 39.6% |
| |||
On-demand treatment (10–60±5 IU/kg) (n=17) | 41.5 | - | |
| |||
Pediatric trial (NCT02210091)37 | Twice-weekly prophylaxis (50±10 IU/kg for 6 months [≥50 EDs]) | 2.0 | 38% |
| |||
BAY 94-9027 (Jivi®) | |||
| |||
PROTECT VIII44 | Twice-weekly prophylaxis (30–40 IU/kg), not eligible for randomisation (n=13) | 4.1 | 15.4% |
| |||
Twice-weekly prophylaxis (30–40 IU/kg), eligible for randomisation (n=11) | 1.9 | 45.5% | |
| |||
Every 5 days prophylaxis (45–60 IU/kg) (n=43) | 1.9 | 44.2% | |
| |||
QW prophylaxis enroled (60 IU/kg) (n=43) | 3.9 | 37.2% | |
| |||
QW prophylaxis completed (60 IU/kg) (n=32) | 1.0 | 50% | |
| |||
On-demand treatment (n=20) | 23.4 | - | |
| |||
PROTECT VIII extension46 | Any PROTECT VIII regimen (n=36) | 1.2c | NR |
| |||
Twice-weekly prophylaxis (n=4) | 1.5 | ||
| |||
Every 5 days prophylaxis (n=10) | 1.1 | ||
| |||
QW prophylaxis (n=8) | 0.4 | ||
|
|||
Variable frequency (n=14) | 2.0 | ||
| |||
N8-GP (Esperoct®) | |||
| |||
Pathfinder 249 | Every 4 days prophylaxis (50 IU/kg) (n=175) | 1.33 | 40% |
| |||
On-demand treatment (n=12) | 30.87 | 0 | |
| |||
Pathfinder 2 extension50 | Every 4 days prophylaxis (50 IU/kg) (n=17) | 0.0 | 52.9% |
| |||
QW prophylaxis (75 IU/kg) (n=38) | 0.0 | 57.9% | |
| |||
Pathfinder 552 | Twice-weekly prophylaxis (50–75 IU/kg) (n=68) | 1.95 | 42.6% |
| |||
Emicizumab (Hemlibra®) | |||
| |||
HAVEN 357 | QW prophylaxis (1.5 mg/kg) (n=36) | 2.5d | 50% |
| |||
Q2W prophylaxis (3.0 mg/kg) (n=35) | 2.6d | 40% | |
| |||
No prophylaxis (n=18) | 47.6d | 0 | |
| |||
HAVEN 458,59 | Q4W prophylaxis (6 mg/kg) (n=41; 5 with inhibitors) | 4.5d,e | 29% |
Data cannot be directly compared between studies due to differences in trial design and patient populations.
Patients with no bleeding episodes during the trial.
Interim data.
Median annualised bleeding rate (ABR) during the full 5-year period.
All bleeding events, regardless of treatment with FVIII.
Median ABR during the expansion phase.
NR: not reported; QW: once weekly; Q2W: every 2 weeks; Q4W: every 4 weeks.
PEGylated extended plasma half-life products
BAX 855 (rurioctocog alpha pegol, Adynovate®, Takeda Pharmaceutical Company Ltd., Lexington, MA, USA), a PEGylated, full-length, recombinant FVIII produced in Chinese hamster ovary (CHO) cells and manufactured by attaching a branched 20 kDa PEG molecule to a SHL rFVIII (Advate®, Takeda US Inc., Lexington, MA, USA)34, was licensed in 2017, with indications for the treatment and prevention of bleeding and perioperative management of haemophilia A35,36. The efficacy, pharmacokinetics and safety of BAX 855 for prophylaxis and on-demand therapy were assessed in previously treated patients with severe haemophilia A in the PROLONG-ATE trial23. The mean half-life of BAX 855 was 1.4-fold longer than that of Advate®. Median ABR in the group assigned to twice-weekly prophylaxis was 1.9 vs 41.5 in the group assigned to on-demand treatment, with no bleeds reported in 39.6% of patients receiving prophylaxis (Table I). No FVIII inhibitors were detected. Furthermore, the efficacy, pharmacokinetics and safety of prophylaxis with BAX 855 were also assessed in previously treated paediatric patients with severe haemophilia A37. The mean half-life of BAX 855 was 1.3–1.5-fold longer than that of Advate®, depending on the FVIII assay method used; the median ABR was 2.0 and no inhibitor was detected. BAX 855 was well tolerated and effective for perioperative use in patients with haemophilia A38.
BAY 94-9027 (Jivi®, Bayer HealthCare Pharmaceuticals, Berlin, Germany), a recombinant EHL FVIII product conjugated with a 60-kDa branched PEG molecule, was licensed in 2018 for previously treated adults and adolescents (aged ≥12 years) for routine prophylaxis, on-demand treatment, and perioperative management of bleeding22,39–42. In a phase I study carried out in patients with severe haemophilia A, this product demonstrated a longer FVIII half-life and higher area under the curve (AUC) than a SHL rFVIII (rFVIII-FS [Kogenate® FS, Bayer, Berkeley, CA, USA])22,43. Furthermore, in a head-to-head pharmacokinetic study in patients with severe haemophilia A, BAY 94-9027 demonstrated improvements in AUC, clearance and half-life compared with the rFVIII-Fc Eloctate® (Swedish Orphan Biovitrum Ltd., Great Abington, UK)25. In the phase II/III PROTECT VIII trial, patients received the product for 36 weeks on demand or prophylactically at individually determined intervals44. Low ABRs were reported in participants across three regimens (median ABRs were 4.1, 1.9 and 3.9 in the twice-weekly, every 5 days and every 7 days interval groups, respectively; zero bleeds occurred in 15.4%, 44.2% and 37.2% of patients, respectively), while median ABR in the on-demand treatment group was 23.4. No participant developed FVIII inhibitors. BAY 94-9027 was also efficacious and well tolerated in the treatment of patients undergoing major surgeries in the PROTECT VIII trial45. The previously demonstrated efficacy and safety of this EHL rFVIII was maintained across the PROTECT VIII extension period: the median ABR was 1.2 and no participant developed FVIII inhibitors46. It must be noted, however, that during the PROTECT VIII Kids trial, a phase III study carried out in 61 previously-treated children aged <12 years with an ongoing extension, 8 children aged 2–6 years left the study because of suspected immunological responses against PEG. No major safety concerns, including FVIII inhibitor development, were observed in these patients47.
N8-GP (turoctocog alfa pegol; Esperoct®, Novo Nordisk, Bagsvaerd, Denmark) is another EHL FVIII product featuring a 40 kDa PEG conjugated to a B-domain truncated FVIII via site-directed glycoPEGylation. It received US Food and Drug Administration (FDA) approval in 2019 and is indicated for use in previously treated adults and children with haemophilia A for prophylaxis, on-demand treatment and the perioperative management of bleeding48. In a phase I pharmacokinetic study (pathfinder 1) in previously treated patients with severe haemophilia A, the product demonstrated a 1.6-fold longer half-life than patients’ previous FVIII products26. The efficacy and safety of N8-GP prophylaxis and on-demand therapy were then assessed in previously treated patients with severe haemophilia A in the pathfinder 2 study49. Patients in the prophylaxis arm had a median ABR of 1.33, 40% of them reporting zero bleeds (Table I). One patient, who developed inhibitory antibodies against FVIII (≥0.6 Bethesda units [BU]) after 93 exposure days with this product, continued into the extension phase of the trial, but was then withdrawn when the FVIII inhibitor titre increased to 13.5 BU. During the pathfinder 2 extension trial, once weekly N8-GP was efficacious in patients with a low bleeding phenotype (≤2 bleeds during the preceding 6 months of the trial’s main phase), with 58% experiencing zero bleeds during the 24-week treatment period (Table I)50. N8-GP was effective and well tolerated for the prevention and treatment of bleeds during major surgery51. Finally, the efficacy, safety and pharmacokinetics of N8-GP prophylaxis and on-demand therapy were assessed in previously treated paediatric patients with severe haemophilia A in the pathfinder 5 trial52. Patients in the prophylaxis arm had a median ABR of 1.95 and 42.6% of them experienced zero bleeds during the study (Table I). No inhibitors were detected.
General comments on extended plasma half-life factor VIII products
These licensed EHL FVIII products were efficacious in previously treated patients with severe haemophilia A, with a median ABR that ranged from 0 to 6.6 in the context of prophylactic regimens (Table I) and compared favourably with values ranging from 18.4 to 41.5 for on-demand regimens. Use of EHL FVIII was accompanied by a lower number and frequency of intravenous infusions than SHL products. However, not all patients were fully protected from bleeding on the dosing regimens tested (twice weekly, every 4 or 5 days or once weekly), which were less frequent than those generally used for prophylaxis with SHL products (thrice weekly or every other day). The ideal target of zero bleeding during the pivotal trials was achieved by approximately half of the patients (Table I), but improvements in ABR during the extension phases suggest that zero bleeds may be achieved for more patients when dosing regimens are individually tailored. There was no increased risk of FVIII inhibitor development in previously treated patients who switched from SHL to EHL FVIII products, but it remains to be seen whether or not this low immunogenicity is replicated in previously untreated patients and in those with a positive family history, i.e. those at higher risk of inhibitor development. Finally, limitations common to all EHL recombinant products are that treatment monitoring through the measurement of plasma FVIII levels may give unreliable results with the widely available one-stage coagulation assays. However, this can be circumvented by employing 2-stage chromogenic assays, although these are more costly and less widely available.
Non-replacement factor therapy: emicizumab
Currently, emicizumab (Hemlibra®, Chugai Pharmaceutical Co., Tokyo, Japan) is the only non-replacement factor therapy licensed for individuals with haemophilia A with or without inhibitors by the FDA and the European Medicines Agency (EMA)53–55. Emicizumab is a bispecific monoclonal antibody that bridges activated factor IX and factor X to mimic the function of missing activated factor VIII, thereby increasing thrombin formation56. The unequivocal advantage of emicizumab for patients without inhibitors is its subcutaneous administration once-weekly (1×W), every 2 weeks (E2W), or every 4 weeks (E4W). However, emicizumab is not suitable for the management of major surgery, nor for treating acute bleeding episodes, because multiple doses are needed in order to achieve a steady state of haemostasis.
The efficacy and safety of emicizumab prophylaxis in patients with severe haemophilia A without inhibitors were assessed in the HAVEN 3 trial57. Patients previously receiving on-demand therapy with FVIII were randomly assigned to prophylaxis with subcutaneous emicizumab at one of two dosing regimens or to no prophylaxis. Participants on 1×W or E2W prophylaxis had lower ABRs for all bleeding events than those with no prophylaxis (2.5, 2.6 vs 47.6, respectively) and a greater percentage had zero bleeding events during the study (50% and 40%, vs 0% with no prophylaxis). In addition, the HAVEN 4 study assessed emicizumab administered E4W in participants with haemophilia A with and without inhibitors58,59. Median ABR for the 41 participants (5 with inhibitors) included in the expansion phase was 4.559.
On the whole, the efficacy results obtained in the multiple HAVEN trials are impressive and there are clear advantages in using the subcutaneous route of administration vs the traditional intravenous route. In spite of a high rate of zero bleeding in the frame of the HAVEN prophylaxis studies, some breakthrough bleeds did require additional haemostatic treatments. Data on the long-term preservation of joint health are currently not available beyond 2 years, and there are some concerns regarding safety. Although thrombotic microangiopathy and thrombosis risk are safety concerns that may be limited to patients with inhibitors treated with high or frequent doses of aPCC60,61, pharmacovigilance is needed to assess the impact on broader populations and over time. In addition, the current difficulty in assaying the haemostatic activity of emicizumab relative to the activity that would be expected from a given level of FVIII is a drawback, both for guiding treatment and recognising inhibitor activity. Indeed, the concomitant use of non-replacement factor therapy with add-on FVIII, e.g. to resolve breakthrough bleeds or manage surgery, requires further investigation to determine whether or not these patients are more likely to develop inhibitors to exogenous FVIII due to limited prior exposure. Finally, whether emicizumab can be used in previously untreated children to delay the use of FVIII replacement and/or to prevent intercurrent bleeding during ITI still needs to be explored. Furthermore, the data from HAVEN do not indicate the level of protection provided by emicizumab prophylaxis in real-world situations, such as in highly active patients and in those who take part in high-impact activities/organised sports62. In addition, continued surveillance is required to establish whether or not the occurrence of anti-drug antibodies, which, in rare cases, neutralise the FVIII-mimicking action of this drug, will be clinically significant.
Discussion and conclusions
Factor VIII replacement remains the standard of care to treat and prevent bleeds in people with haemophilia A, with the unequivocal evidence supported by decades of clinical and real-world data that FVIII prophylaxis is highly effective in reducing bleeding and long-term complications such as arthropathy. It is also evident that FVIII prophylaxis regimens should be tailored to meet the individual needs and expectations of the patients63. The following factors should be considered when individualising a prophylactic regimen: bleeding pattern, joint health, individual pharmacokinetic profile, physical activity, and likelihood of adherence. Recent developments, such as EHL FVIII products and pharmacokinetic-guided dosing, provide new opportunities to optimise regimes for each individual patient64.
So far, in adolescents or adults, no major safety issues have been identified with any of the technologies used to extend the plasma half-life of FVIII65. However, the available data were obtained in the frame of clinical trials enroling highly selected patients, chosen with the goal of demonstrating safety and efficacy, i.e. the end points required for product licensing. Thus, long-term post-marketing surveillance will be extremely important in order to confirm the safety of EHL products when used more extensively and for longer periods of time in real-world situations. For instance, PEGylation is being safely and widely used to prolong the plasma half-life of various therapeutic agents, including epoetin beta, growth hormone, interferon α-2a and anti-tumour necrosis factor monoclonal antibodies66. However, none of these products are being administered lifelong from birth, even though the concentrations of PEG employed to prolong the plasma half-life of FVIII products are very low and, indeed, among the lowest used in currently available pegylated medications67.
Extended half-life products have not raised any concerns about the possibility of an increased risk of FVIII inhibitor formation, at least in previously treated patients who are tolerant to FVIII and thus at low risk of developing this complication. However, the immunogenicity of EHL products compared with SHL products in patients at high risk for inhibitor development, e.g. previously untreated or minimally treated children, and those with a family history of inhibitor, remains to be established.
Taken together, strict and long-term post-marketing surveillance should be put in place and should be supervised independently. This is essential in order to monitor the long-term safety of EHL FVIII products and of non-replacement products such as emicizumab, given that its FVIII-mimicking activity differs from that of FVIII, a protein that plays important physiological roles beyond haemostasis, including in cardiovascular and skeletal health11,56,68. For example, FVIII deficiency is associated with altered bone metabolism, as indicated by reduced formation and increased reabsorption of bone observed in FVIII-deficient mice, and osteoporosis is a common comorbidity in patients with FVIII deficiency69,70. Data are needed to provide information as to whether or not the FVIII-mimicking activity of emicizumab can fulfil the roles of FVIII beyond haemostasis in patients with haemophilia.
Acknowledgements
Editorial assistance was provided by Graeme Baldwin of Darwin Medical Communications (London, UK), funded by Bayer. He produced a summary of the literature, extracted data and assisted with formatting and references. However, all the critical statements on the therapeutic products featured in this article are written with the full and independent responsibility of all the Authors, who contributed equally to the drafting, editing and final version of this manuscript.
Footnotes
Disclosures of conflicts of interest
LA has participated in a Data Safety Monitoring Board for Octapharma and Takeda. He has also received honoraria from Bayer, Kedrion, and Takeda, and has acted as Chair for a scientific advisory committee for Kedrion. PMM sits on the scientific board for the Bayer Awards. He has also received honoraria for lectures at educational symposia from Bayer, Kedrion and Novo Nordisk. WS has participated in a Data Safety Monitoring Board for Biotest and received honoraria for lectures from Biotest, Novo Nordisk, Roche, Takeda and Bio&Bio. MT has been an advisor for Amgen, Genentech, Kedrion, Novo Nordisk, Octapharma and Takeda. He has received fees for lectures at symposia for Takeda. Research with Novo Nordisk and Takeda.
References
- 1.Mannucci PM, Iacobelli M. Progress in the contemporary management of hemophilia: The new issue of patient aging. Eur J Intern Med. 2017;43:16–21. doi: 10.1016/j.ejim.2017.05.012. [DOI] [PubMed] [Google Scholar]
- 2.Chen SL. Economic costs of hemophilia and the impact of prophylactic treatment on patient management. Am J Manag Care. 2016;22:s126–33. [PubMed] [Google Scholar]
- 3.Wight J, Paisley S. The epidemiology of inhibitors in haemophilia A: a systematic review. Haemophilia. 2003;9:418–35. doi: 10.1046/j.1365-2516.2003.00780.x. [DOI] [PubMed] [Google Scholar]
- 4.Rocino A, Franchini M, Coppola A. treatment and prevention of bleeds in haemophilia patients with inhibitors to factor VIII/IX. J Clin Med. 2017;6:e46. doi: 10.3390/jcm6040046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.DiMichele DM, Hoots WK, Pipe SW, et al. International workshop on immune tolerance induction: consensus recommendations. Haemophilia. 2007;13:1–22. doi: 10.1111/j.1365-2516.2007.01497.x. [DOI] [PubMed] [Google Scholar]
- 6.Hay CR, DiMichele DM International Immune Tolerance S. The principal results of the International Immune Tolerance Study: a randomized dose comparison. Blood. 2012;119:1335–44. doi: 10.1182/blood-2011-08-369132. [DOI] [PubMed] [Google Scholar]
- 7.Zanon E, Milan M, Pasca S. Immune tolerance induction rescue with turoctocog-alfa in a poor risk haemophilia A inhibitor young child: the history of a success. Blood Coagul Fibrinolysis. 2018;29:465–68. doi: 10.1097/MBC.0000000000000736. [DOI] [PubMed] [Google Scholar]
- 8.National Health Service (NHS) England [internet] Clinical Commissioning Policy: immune tolerance induction (ITI) for haemophilia A (all ages) [Accessed on 06/02/2019]. Available at: https://www.england.nhs.uk/wp-content/uploads/2018/07/Immune-tolerance-induction-for-haemophilia-A-all-ages.pdf.
- 9.Manco-Johnson MJ, Abshire TC, Shapiro AD, et al. Prophylaxis versus episodic treatment to prevent joint disease in boys with severe hemophilia. N Engl J Med. 2007;357:535–44. doi: 10.1056/NEJMoa067659. [DOI] [PubMed] [Google Scholar]
- 10.Gringeri A, Lundin B, von Mackensen S, et al. ESPRIT Study Group. A randomized clinical trial of prophylaxis in children with hemophilia A (the ESPRIT Study) J Thromb Haemost. 2011;9:700–10. doi: 10.1111/j.1538-7836.2011.04214.x. [DOI] [PubMed] [Google Scholar]
- 11.Samuelson Bannow B, Recht M, Negrier C, et al. Factor VIII: long-established role in haemophilia A and emerging evidence beyond haemostasis. Blood Rev. 2019;35:43–50. doi: 10.1016/j.blre.2019.03.002. [DOI] [PubMed] [Google Scholar]
- 12.Hoots WK. Emergency care issues in hemophilia. Treatment of hemophilia. 2007. [Accessed on 06/02/2019]. p. 43. Available at: http://www1.wfh.org/publication/files/pdf-1196.pdf.
- 13.Andersson NG, Auerswald G, Barnes C, et al. Intracranial haemorrhage in children and adolescents with severe haemophilia A or B - the impact of prophylactic treatment. Br J Haematol. 2017;179:298–307. doi: 10.1111/bjh.14844. [DOI] [PubMed] [Google Scholar]
- 14.Aledort LM, Haschmeyer RH, Pettersson H. A longitudinal study of orthopaedic outcomes for severe factor-VIII-deficient haemophiliacs. The Orthopaedic Outcome Study Group. J Intern Med. 1994;236:391–9. doi: 10.1111/j.1365-2796.1994.tb00815.x. [DOI] [PubMed] [Google Scholar]
- 15.Valentino LA, Mamonov V, Hellmann A, et al. A randomized comparison of two prophylaxis regimens and a paired comparison of on-demand and prophylaxis treatments in hemophilia A management. J Thromb Haemost. 2012;10:359–67. doi: 10.1111/j.1538-7836.2011.04611.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Oladapo AO, Epstein JD, Williams E, et al. Health-related quality of life assessment in haemophilia patients on prophylaxis therapy: a systematic review of results from prospective clinical trials. Haemophilia. 2015;21:e344–58. doi: 10.1111/hae.12759. [DOI] [PubMed] [Google Scholar]
- 17.Den Uijl IE, Mauser Bunschoten EP, Roosendaal G, et al. Clinical severity of haemophilia A: does the classification of the 1950s still stand? Haemophilia. 2011;17:849–53. doi: 10.1111/j.1365-2516.2011.02539.x. [DOI] [PubMed] [Google Scholar]
- 18.Iorio A. Using pharmacokinetics to individualize hemophilia therapy. Hematology Am Soc Hematol Educ Program. 2017;2017:595–604. doi: 10.1182/asheducation-2017.1.595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.von Mackensen S, Kalnins W, Krucker J, et al. Haemophilia patients’ unmet needs and their expectations of the new extended half-life factor concentrates. Haemophilia. 2017;23:566–74. doi: 10.1111/hae.13221. [DOI] [PubMed] [Google Scholar]
- 20.Carcao MD, Aledort L. Prophylactic factor replacement in hemophilia. Blood Rev. 2004;18:101–13. doi: 10.1016/S0268-960X(03)00038-9. [DOI] [PubMed] [Google Scholar]
- 21.Saxena K. Barriers and perceived limitations to early treatment of hemophilia. J Blood Med. 2013;4:49–56. doi: 10.2147/JBM.S43734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Coyle TE, Reding MT, Lin JC, et al. Phase I study of BAY 94–9027, a PEGylated B-domain-deleted recombinant factor VIII with an extended half-life, in subjects with hemophilia A. J Thromb Haemost. 2014;12:488–96. doi: 10.1111/jth.12506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Konkle BA, Stasyshyn O, Chowdary P, et al. Pegylated, full-length, recombinant factor VIII for prophylactic and on-demand treatment of severe hemophilia A. Blood. 2015;126:1078–85. doi: 10.1182/blood-2015-03-630897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Powell JS, Josephson NC, Quon D, et al. Safety and prolonged activity of recombinant factor VIII Fc fusion protein in hemophilia A patients. Blood. 2012;119:3031–7. doi: 10.1182/blood-2011-09-382846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Shah A, Solms A, Wiegemann S, et al. BAY 94–9027 and recombinant factor VIII Fc fusion protein: a head-to-head, randomized, crossover, pharmacokinetic study in patients with severe haemophilia A. Haemophilia. 2019;25:45. [Google Scholar]
- 26.Tiede A, Brand B, Fischer R, et al. Enhancing the pharmacokinetic properties of recombinant factor VIII: first-in-human trial of glycoPEGylated recombinant factor VIII in patients with hemophilia A. J Thromb Haemost. 2013;11:670–8. doi: 10.1111/jth.12161. [DOI] [PubMed] [Google Scholar]
- 27.US Food and Drug Administration [internet] ELOCTATE prescribing information. 2017. [Accessed on 06/02/2019]. Available at: https://www.fda.gov/downloads/BiologicsBloodVaccines/BloodBloodProducts/ApprovedProducts/LicensedProductsBLAs/FractionatedPlasmaProducts/UCM400192.pdf.
- 28.European Medicines Agency [internet] ELOCTA Summary of Product Characteristics. 2015. [Accessed on 06/02/2019]. Available at: https://www.ema.europa.eu/documents/product-information/elocta-epar-product-information_en.pdf.
- 29.Mahlangu J, Powell JS, Ragni MV, et al. Phase 3 study of recombinant factor VIII Fc fusion protein in severe hemophilia A. Blood. 2014;123:317–25. doi: 10.1182/blood-2013-10-529974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Young G, Mahlangu J, Kulkarni R, et al. Recombinant factor VIII Fc fusion protein for the prevention and treatment of bleeding in children with severe hemophilia A. J Thromb Haemost. 2015;13:967–77. doi: 10.1111/jth.12911. [DOI] [PubMed] [Google Scholar]
- 31.Nolan B, Mahlangu J, Perry D, et al. Long-term safety and efficacy of recombinant factor VIII Fc fusion protein (rFVIIIFc) in subjects with haemophilia A. Haemophilia. 2016;22:72–80. doi: 10.1111/hae.12766. [DOI] [PubMed] [Google Scholar]
- 32.Mahlangu JN, Ragni M, Gupta N, et al. Long-acting recombinant factor VIII Fc fusion protein (rFVIIIFc) for perioperative haemostatic management in severe haemophilia A. Thromb Haemost. 2016;116:1–8. doi: 10.1160/TH15-10-0780. [DOI] [PubMed] [Google Scholar]
- 33.Carcao M, Shapiro A, Staber JM, et al. Recombinant factor VIII Fc fusion protein for immune tolerance induction in patients with severe haemophilia A with inhibitors-A retrospective analysis. Haemophilia. 2018;24:245–52. doi: 10.1111/hae.13413. [DOI] [PubMed] [Google Scholar]
- 34.Turecek PL, Bossard MJ, Graninger M, et al. BAX 855, a PEGylated rFVIII product with prolonged half-life. Development, functional and structural characterisation. Hämostaseologie. 2012;32:S29–38. [PubMed] [Google Scholar]
- 35.US Food and Drug Administration [internet] ADYNOVATE prescribing information. 2016. [Accessed on 06/02/2019]. Available at: https://www.fda.gov/downloads/BiologicsBloodVaccines/BloodBloodProducts/ApprovedProducts/LicensedProductsBLAs/FractionatedPlasmaProducts/UCM472594.pdf.
- 36.European Medicines Agency [internet] ADYNOVI summary of product characteristics. 2018. [Accessed on 06/02/2019]. Available at: https://www.ema.europa.eu/documents/product-information/adynovi-epar-product-information_en.pdf.
- 37.Mullins ES, Stasyshyn O, Alvarez-Roman MT, et al. Extended half-life pegylated, full-length recombinant factor VIII for prophylaxis in children with severe haemophilia A. Haemophilia. 2017;23:238–46. doi: 10.1111/hae.13119. [DOI] [PubMed] [Google Scholar]
- 38.Gruppo R, Lopez-Fernandez MF, Wynn TT, et al. Perioperative haemostasis with full-length, PEGylated, recombinant factor VIII with extended half-life (rurioctocog alfa pegol) in patients with haemophilia A: final results of a multicentre, single-arm phase III trial. Haemophilia. 2019;25:773–81. doi: 10.1111/hae.13807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.US Food and Drug Administration [internet] JIVI Antihemophilic Factor (Recombinant) PEGylated-aucl prescribing information. 2018. [Accessed on 11/01/2019]. Available at: https://www.fda.gov/downloads/BiologicsBloodVaccines/UCM618979.pdf.
- 40.European Medicines Agency [internet] JIVI Summary of product characteristics. 2018. [Accessed on 11/01/2019]. Available at: https://www.ema.europa.eu/documents/product-information/jivi-epar-product-information_en.pdf.
- 41.Mei B, Pan C, Jiang H, et al. Rational design of a fully active, long-acting PEGylated factor VIII for hemophilia A treatment. Blood. 2010;116:270–9. doi: 10.1182/blood-2009-11-254755. [DOI] [PubMed] [Google Scholar]
- 42.Pharmaceuticals and Medical Devices Agency [internet] Report on the Deliberation Results. 2018. [Accessed on 05/11/2019]. Available at: http://www.pmda.go.jp/files/000229786.pdf.
- 43.Shah A, Coyle T, Lalezari S, et al. BAY 94–9027, a PEGylated recombinant factor VIII, exhibits a prolonged half-life and higher area under the curve in patients with severe haemophilia A: comprehensive pharmacokinetic assessment from clinical studies. Haemophilia. 2018;24:733–40. doi: 10.1111/hae.13561. [DOI] [PubMed] [Google Scholar]
- 44.Reding MT, Ng HJ, Poulsen LH, et al. Safety and efficacy of BAY 94–9027, a prolonged-half-life factor VIII. J Thromb Haemost. 2017;15:411–19. doi: 10.1111/jth.13597. [DOI] [PubMed] [Google Scholar]
- 45.Santagostino E, Lalezari S, Reding MT, et al. Safety and efficacy of BAY 94–9027, an extended-half-life factor VIII, during surgery in patients with severe hemophilia A: Results of the PROTECT VIII clinical trial. Thromb Res. 2019;183:13–19. doi: 10.1016/j.thromres.2019.08.023. [DOI] [PubMed] [Google Scholar]
- 46.Reding MT, Poulsen LH, Andre Holme P, et al. Efficacy and safety of BAY 94-9027 is sustained for 5 years: outcomes in 36 patients in the PROTECT VIII extension study. Res Prac Thromb Haemost. 2019;3:122. [Google Scholar]
- 47.Santagostino E, Saxena K, Kenet G, et al. PROTECT VIII Kids trial results: BAY 94–9027 safety and efficacy in previously treated children with severe hemophilia A. Haemophilia. 2016;22:41. doi: 10.1111/hae.13963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.US Food and Drug Administration [internet] Approval Letter - ESPEROCT. BL 125671/0. Feb 19, 2019. [Accessed on 10/10/2019]. Available at: https://www.fda.gov/media/120350/download.
- 49.Giangrande P, Andreeva T, Chowdary P, et al. Clinical evaluation of glycoPEGylated recombinant FVIII: efficacy and safety in severe haemophilia A. Thromb Haemost. 2017;117:252–61. doi: 10.1160/TH16-06-0444. [DOI] [PubMed] [Google Scholar]
- 50.Curry N, Albayrak C, Escobar M, et al. Once-weekly prophylaxis with glycoPEGylated recombinant factor VIII (N8-GP) in severe haemophilia A: safety and efficacy results from pathfinder 2 (randomized phase III trial) Haemophilia. 2019;25:373–81. doi: 10.1111/hae.13712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hampton K, Chowdary P, Dunkley S, et al. First report on the safety and efficacy of an extended half-life glycoPEGylated recombinant FVIII for major surgery in severe haemophilia A. Haemophilia. 2017;23:689–96. doi: 10.1111/hae.13246. [DOI] [PubMed] [Google Scholar]
- 52.Meunier S, Alamelu J, Ehrenforth S, et al. Safety and efficacy of a glycoPEGylated rFVIII (turoctocog alpha pegol, N8-GP) in paediatric patients with severe haemophilia A. Thromb Haemost. 2017;117:1705–13. doi: 10.1160/TH17-03-0166. [DOI] [PubMed] [Google Scholar]
- 53.European Medicines Agency [internet] CHMP post-authorisation summary of positive opinion for Hemlibra (II-02) [Accessed on 06/02/2019]. Available at: https://www.ema.europa.eu/en/documents/smop/chmp-post-authorisation-summary-positive-opinion-hemlibra-ii-02_en.pdf.
- 54.European Medicines Agency [internet] HEMLIBRA summary of product characteristics. 2018. [Accessed on 06/02/2019]. Available at: https://www.ema.europa.eu/documents/product-information/hemlibra-epar-product-information_en.pdf.
- 55.US Food and Drug Administration [internet] HEMLIBRA prescribing information. 2018. [Accessed on 06/02/2019]. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/761083s002s004lbl.pdf.
- 56.Kitazawa T, Igawa T, Sampei Z, et al. A bispecific antibody to factors IXa and X restores factor VIII hemostatic activity in a hemophilia A model. Nat Med. 2012;18:1570–4. doi: 10.1038/nm.2942. [DOI] [PubMed] [Google Scholar]
- 57.Mahlangu J, Oldenburg J, Paz-Priel I, et al. Emicizumab prophylaxis in patients who have hemophilia A without inhibitors. N Engl J Med. 2018;379:811–22. doi: 10.1056/NEJMoa1803550. [DOI] [PubMed] [Google Scholar]
- 58.Jiménez-Yuste V, Shima M, Fukutake K, et al. Emicizumab subcutaneous dosing every 4 weeks for the management of hemophilia A: preliminary data from the pharmacokinetic run-in cohort of a multicenter, Open-Label, Phase 3 Study (HAVEN 4) Blood. 2017;130(Suppl 1):86. [Google Scholar]
- 59.Pipe SW, Shima M, Lehle M, et al. Efficacy, safety, and pharmacokinetics of emicizumab prophylaxis given every 4 weeks in people with haemophilia A (HAVEN 4): a multicentre, open-label, non-randomised phase 3 study. Lancet Haematol. 2019;6:e295–305. doi: 10.1016/S2352-3026(19)30054-7. [DOI] [PubMed] [Google Scholar]
- 60.Oldenburg J, Mahlangu JN, Kim B, et al. Emicizumab prophylaxis in hemophilia A with inhibitors. N Engl J Med. 2017;377:809–18. doi: 10.1056/NEJMoa1703068. [DOI] [PubMed] [Google Scholar]
- 61.Genentech [internet] HEMLIBRA® (emicizumab-kxwh) for US Patients and Caregivers. Reported fatalities. [Accessed on 10/10/2019]. Available at: https://www.emicizumabinfo.com/content/dam/gene/emicizumabinfo/patient/pdfs/reported-fatalities.pdf.
- 62.Medical and Scientific Advisory Council. Recommendation on the Use and Management of Emicizumab-kxwh (Hemlibra®) for Hemophilia A with and without Inhibitors. 2018. [Accessed on 06/06/2019]. Available at: https://www.hemophilia.org/Researchers-Healthcare-Providers/Medical-and-Scientific-Advisory-Council-MASAC/MASAC-Recommendations/Recommendation-on-the-Use-and-Management-of-Emicizumab-kxwh-Hemlibra-for-Hemophilia-A-with-and-without-Inhibitors.
- 63.Petrini P, Valentino LA, Gringeri A, et al. Individualizing prophylaxis in hemophilia: a review. Expert Rev Hematol. 2015;8:237–46. doi: 10.1586/17474086.2015.1002465. [DOI] [PubMed] [Google Scholar]
- 64.Langer AL, Etra A, Aledort L. Evaluating the safety of emicizumab in patients with hemophilia A. Expert Opin Drug Saf. 2018;17:1233–37. doi: 10.1080/14740338.2019.1551356. [DOI] [PubMed] [Google Scholar]
- 65.Mancuso ME, Santagostino E. Outcome of clinical trials with new extended half-life FVIII/IX concentrates. J Clin Med. 2017;6 doi: 10.3390/jcm6040039. pii: E39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ivens IA, Baumann A, McDonald TA, et al. PEGylated therapeutic proteins for haemophilia treatment: a review for haemophilia caregivers. Haemophilia. 2013;19:11–20. doi: 10.1111/j.1365-2516.2012.02931.x. [DOI] [PubMed] [Google Scholar]
- 67.Baumann A, Piel I, Hucke F, et al. Pharmacokinetics, excretion, distribution, and metabolism of 60-kDa polyethylene glycol used in BAY 94–9027 in rats and its value for human prediction. Eur J Pharm Sci. 2019;130:11–20. doi: 10.1016/j.ejps.2019.01.015. [DOI] [PubMed] [Google Scholar]
- 68.Lenting PJ, Denis CV, Christophe OD. Emicizumab, a bispecific antibody recognizing coagulation factors IX and X: how does it actually compare to factor VIII? Blood. 2017;130:2463–68. doi: 10.1182/blood-2017-08-801662. [DOI] [PubMed] [Google Scholar]
- 69.Kiper Unal HD, Comert Ozkan M, Atilla FD, et al. Evaluation of bone mineral density and related parameters in patients with haemophilia: a single center cross-sectional study. Am J Blood Res. 2017;7:59–66. [PMC free article] [PubMed] [Google Scholar]
- 70.Weitzmann MN, Roser-Page S, Vikulina T, et al. Reduced bone formation in males and increased bone resorption in females drive bone loss in hemophilia A mice. Blood Adv. 2019;3:288–300. doi: 10.1182/bloodadvances.2018027557. [DOI] [PMC free article] [PubMed] [Google Scholar]