Subscribe to RSS
DOI: 10.1055/s-0042-1756188
The More Recent History of Hemophilia Treatment
- Abstract
- Search Strategy
- Replacement Therapy: from Standard to Extended Half-Life Recombinant Products
- Management of Inhibitor Patients and Non-replacement Therapy
- Gene Therapy
- Conclusion
- References
Abstract
The availability first in the 1970s of plasma-derived and then in the 1990s of recombinant clotting factor concentrates represented a milestone in hemophilia care, enabling not only treatment of episodic bleeding events but also implementation of prophylactic regimens. The treatment of hemophilia has recently reached new landmarks. The traditional clotting factor replacement therapy for hemophilia has been substituted over the last 10 years by novel treatments such as bioengineered factor VIII and IX molecules with extended half-life and non-factor treatments including the bispecific antibody emicizumab. This narrative review is dedicated to these newer therapies, which are contributing significantly to improving the long-term management of prophylaxis in hemophilia patients. Another section is focused on the current state of gene therapy, which is a promising definitive cure for severe hemophilia A and B.
#
The hemophilias are X chromosome-linked bleeding disorders, resulting from the deficiency or dysfunction of coagulation factor VIII (FVIII, hemophilia A) or factor IX (FIX, hemophilia B).[1] These rare inherited disorders (the prevalence of hemophilia A and B is 1:5,000 and 1:30,000 male live births, respectively) and are clinically relevant, with symptoms being loosely commensurate with the degree of plasma factor deficiency.[1] [2] People with hemophilia are at risk of spontaneous bleeding, and trauma and surgical interventions can provoke uncontrolled bleeding.[3] [4] [5] The main sites of spontaneous bleeding are joints and muscles; if these bleeds are not treated adequately, chronic damage to the musculoskeletal system can ensue, with consequent severe handicap and disability.
An early reference to a bleeding condition highly suggestive of hemophilia dates back to the second century AD. The Babylonian Talmud exempted male infants from circumcision if they had two brothers who had already died of excessive bleeding from the procedure. Albucasis, an Arabic physician who lived in the 10th century, described a family with males who died from bleeding after minor injuries. The first modern description of hemophilia appears to be that by Dr. John Conrad Otto, a physician from Philadelphia, who in 1803 described an inherited bleeding disorder in several families in which only males, called bleeders, were affected, and in which transmission occurred via unaffected females. The word “hemophilia” appeared, however, for the first time in an essay written in 1828 by the German physician Johann Lukas Schonlein and his student Friedrich Hopff. Hemophilia B was distinguished from the more common hemophilia A in 1952, and was often initially denoted as Christmas disease after the surname of the first boy described with the condition. The first description of the genetics of hemophilia was published in 1820 by Nasse.[6] [7] Hemophilia is occasionally called “the royal disease”, because several members of royal families across the whole of Europe were affected by this affliction, which they inherited from Victoria, Queen of England from 1837 to 1901, who was a hemophilia B carrier.[8] Queen Victoria's eighth son Leopold had hemophilia B, suffered from frequent bleeds and died of a brain hemorrhage at the age of 31. Two of her daughters, Alice and Beatrice, were carriers of hemophilia B and transmitted the disease to the Spanish, German and Russian royal families.
In the 1950s and early 1960s, people with hemophilia could only be treated with whole blood or fresh plasma. Unfortunately, there is insufficient FVIII or FIX in these blood products to arrest severe bleeding. Thus, most people with severe hemophilia died in childhood or early adulthood of bleeding after surgery or trauma or hemorrhages in vital organs (particularly the brain).[9] [10] The patients who survived developed severe damage to the musculoskeletal system, which resulted in them being bedridden or confined to a wheelchair. In 1964, Judith Graham Pool discovered that the precipitate left from thawing and spinning plasma contained large amounts of FVIII. This was a huge step forward in hemophilia care since it enabled blood banks to produce and store cryoprecipitate and, for the first time, enough FVIII could be infused in relatively small volumes to control severe bleeding and to make emergency and elective surgery possible.[11]
The most significant advance in hemophilia therapy occurred in the 1970s, due to the industrial manufacturing and large-scale commercial availability of freeze-dried plasma concentrates containing FVIII and FIX. This innovation revolutionized hemophilia care because factor concentrates could be stored easily, which made it possible to infuse these products at home. The widespread adoption of home therapy enabled early control of bleeds with a related decrease in musculoskeletal damage and a drastic reduction in the requirement of hospital visits, resulting in dramatic improvements of quality of life and life expectancy.[10] Sweden was a pioneer in using such products to implement the prophylactic treatment of hemorrhages (primary prophylaxis), instead of only treating bleeding, episodically, when it occurred.[12] [13] In 1977, the demonstration that desmopressin (DDVP), a synthetic drug, was efficacious clinically as a non-transfusional method of FVIII replacement in mild hemophilia A and von Willebrand disease (VWD) was another important step forward.[14] A major advantage of DDAVP was that of reducing patients' exposure to large-pool, plasma-derived products which, unfortunately, had been responsible for the transmission of blood-borne viruses (the human immunodeficiency virus and the hepatitis B and C viruses) during 1980s.[15] However, this gloomy decade was accompanied by progresses in virucidal or viral removal techniques, which greatly increased the safety of plasma-derived coagulation products, and molecular medicine, which led to the first production of recombinant coagulation FVIII and IX in the 1990s.[16] The narration of this review begins with this period, which is conventionally considered the beginning of modern hemophilia therapy. This review is an update of a previous one on the history of hemophilia published to celebrate the 40th anniversary of this Journal.[6] [Fig. 1] summarizes the most important progresses, decade by decade, of hemophilia therapy.
Search Strategy
For this narrative review we analyzed the medical literature for published articles on the recent history of hemophilia therapy. The Medline and PubMed electronic database was searched for publications without temporal limits using English language as a restriction. The Medical Subject Heading and keywords used were: “hemophilia A,” “hemophilia B,” “therapy,” “history,” “prophylaxis,” “recombinant coagulation products,” “plasma-derived concentrates,” “standard,” “extended half-life,” “bleeding,” “inhibitor,” “replacement therapy,” “non-replacement therapy,” “emicizumab,” “gene therapy,” and “quality of life.” We also screened the reference lists of the studies identified and of the most relevant review articles for additional studies not captured in our initial literature search.
#
Replacement Therapy: from Standard to Extended Half-Life Recombinant Products
As mentioned, the prevention of bleeding through prophylaxis has become the evidence-based standard of care over the last 20 years after it was demonstrated to be superior to on-demand management of bleeds in preserving the articular integrity of hemophilia patients.[17] [18] In addition, the availability of large amounts of increasingly safe recombinant clotting factor concentrates (i.e., manufactured with ultrafiltration and nanofiltration viral inactivation techniques and with no proteins other than FVIII and FIX in the culture medium or final formulation) allowed a widespread implementation of prophylaxis regimens, at least in high-income countries.[19] However, while primary prophylaxis has important advantages, with a dramatic improvement of patients' quality of life, it also has have some drawbacks, such as the need of frequent intravenous injections owing to the short plasma half-life of the replaced coagulation factors (range 10–12 hours for FVIII, 18–20 hours for FIX). This problem can lead to suboptimal adherence to prophylaxis, particularly by younger patients in whom the issue of vein access is particularly cogent, such that it is sometimes necessary to use ports or other central venous access devices for the delivery of replacement therapy.
Starting in 2010s, attempts were made to overcome this problem by engineering clotting factors using recombinant technology, the goal being to obtain therapeutic products that, through higher factor peaks and trough levels, remained longer in the circulation and therefore managed to reduce the frequency of intravenous injections and the burden of prophylaxis.[20] The two techniques mainly adopted were coagulation factor fusion to proteins such as the fragment crystallizable (Fc) component of IgG1 or albumin; and conjugation with chemicals such as polyethylene glycol (PEG).[21] [22] [23] While albumin and Fc fusion prolong coagulation factor plasma half-life through avoidance of clearance by endolysosomal degradation, PEG, which is attached site-specifically or randomly to coagulation factors, acts by reducing clotting factor susceptibility to proteolysis and renal elimination.[24] [25] [26] A number of extended half-life (EHL) recombinant FVIII and FIX coagulation factors have been licensed and marketed over the last 8 years ([Table 1]). The strategies to improve pharmacokinetic parameters resulted in significant extensions of the half-life of FIX concentrates, usually three to five times longer than the standard half-life (SHL) of standard FIX products.[27] [28] [29] [30] [31] [32] However, EHL recombinant FVIII products achieved half-lives only 1.5 to 1.7 times longer than those of SHL FVIII concentrates.[33] [34] [35] [36] [37] [38] [39] [40] [41] [42] This lesser improvement in pharmacokinetic parameters is due to the binding of FVIII to von Willebrand factor (VWF), which stabilizes this moiety in plasma. Consequently, the maximum half-life that can be achieved by EHL FVIII products is the same as that of VWF.[43] A novel EHL recombinant FVIII product (BIVV001), created by the fusion of the B-domain deleted FVIII protein with the D′D3 domain of VWF, is under clinical development with the goal to overcome this limitation.[44]
Abbreviations: rFIX, recombinant factor IX; rFVIII, recombinant factor VIII; SHL, standard half-life.
Source: Adapted from Mannucci, 2020[20] and Ozelo and Yamaguti-Hayakawa, 2022.[48]
Finally, improved VWF affinity and prolongation of the FVIII half-life were also recently attempted by using the single chain technology, in which the heavy and light chains of FVIII are covalently bound together to form a novel, more stable, recombinant protein.[45]
Pivotal clinical studies on adults and children with hemophilia identified EHL products as effective and safe in managing surgical interventions as well as in stopping or preventing bleeding in the context of episodic and prophylactic treatment regimens, with median annualized bleeding rates being markedly lower in patients receiving prophylaxis than in those receiving episodic treatment.[26] [46] [47] [48] Thanks to their better pharmacokinetic profile, EHL FVIII products can be administered effectively prophylactically twice a week instead of three times a week as done for SHL FVIII products. EHL FIX products are, however, much more satisfactory, because they can be administered every 10 or even 15 days. From a clinical point of view, the higher trough levels of both FVIII (2–3%) and FIX (5–10%) that can be achieved with EHL than with SHL products have lowered the annual burden of intravenous injections (average reduction of approximately 60% with EHL FIX and 30% with EHL FVIII) and hence decreased the annual consumption in units of EHL products, while maintaining a favorable median annualized bleeding rate. All these factors result in an improvement of adherence to prophylaxis and, ultimately, a better quality of life for patients.[24]
#
Management of Inhibitor Patients and Non-replacement Therapy
The development of inhibitory alloantibodies against FVIII or FIX is one of the most challenging complications of replacement therapy. Inhibitors, which develop in approximately 30% of patients with severe hemophilia A and in 10% of those with severe hemophilia B, make replacement therapy ineffective, limit patients' access to a safe and effective prophylaxis and increase their risks of morbidity and mortality.[49] [50] [51] [52] The rate of inhibitor formation varies depending on various factors, including the degree of deficiency, ethnicity (hemophiliacs belonging to African or Hispanic races have higher risks of inhibitor formation), type of genetic mutations (complete deletion of F8 or F9 and other null gene mutations predispose to inhibitor development) and environmental factors, the last including the class of replacement product, with plasma-derived products being associated with a reduced risk of inhibitors compared with recombinant products.[53] Although the introduction of bypassing agents, including recombinant activated factor VII (rFVIIa, NovoSeven) and activated prothrombin complex concentrates (Factor Eight Inhibitor Bypassing Activity or FEIBA), in the 1990s was a major improvement in the management of acute bleeding in inhibitor patients, several unmet needs still remained, first of all to reduce the frequency of the intravenous injections and render the prophylactic regimens more practical.[54] [55] [56] [57] Considering these unmet needs, innovative therapeutic strategies were developed not based on replacing or bypassing the deficient factor.[58] [59] Such non-factor-based therapies act by amplifying the coagulation cascade to generate thrombin (emicizumab) or by inhibiting naturally occurring anticoagulant pathways (fitusiran and concizumab).[59] So far, however, only the bispecific monoclonal antibody emicizumab has been licensed and marketed.[52] Emicizumab, by aligning activated FIX and factor X, facilitates their spatial interaction and thus promotes thrombin formation by mimicking the cofactor activity of activated FVIII independently of the degree of factor deficiency and the presence of inhibitors. Administered subcutaneously, emicizumab concentrations reach a steady state with a long plasma half-life, enabling long dosing intervals (at least every week or even every 2 weeks).[60] [61] [62] A series of pivotal trials (HAVEN 1-4) confirmed the efficacy and safety of emicizumab for prophylaxis of bleeding in children and adults with hemophilia A, with and without inhibitors, and this drug is currently licensed with these indications.[63] [64] [65] [66] The high rate of zero bleeding observed in the HAVEN trials of prophylaxis makes emicizumab the first-choice therapy for regular prophylaxis in hemophilia A patients with inhibitors. The fact that it has also been licensed for hemophilia A patients without inhibitors makes it an important alternative to current available options of SHL and EHL clotting factors, and it also has the advantage of a subcutaneous rather than intravenous route of administration, although no head-to-head comparative studies between replacement and non-replacement products have been conducted. Various randomized trials are, however, assessing the use of emicizumab prophylaxis in previously untreated children with severe hemophilia A at high risk of developing inhibitors.[48] Apart from the evident advantages of emicizumab, first and foremost the user-friendly subcutaneous route of administration which is particularly attractive for infants and children, a number of its potential disadvantages must be considered, including the difficulty in laboratory monitoring as proxy of its hemostatic efficacy and the still unproven long-term impact on the preservation of joint and bone heath.[24] [67] In addition, the management of breakthrough bleeds or surgery in severe hemophilia A patients under emicizumab prophylaxis may be challenging.[68] [69] In this context, the concomitant use of activated prothrombin complex concentrates should be avoided and rFVIIa preferred in inhibitor patients due to the lower thrombotic risk of the latter.[69] Similarly, the risk of de novo inhibitor development arising from the use of FVIII replacement in previously untreated patients in dangerous, high-risk circumstances (i.e., trauma, surgery) warrants more data.[48] Finally, although it has been hypothesized that the use of emicizumab prophylaxis during immune tolerance induction (ITI) in order to reduce bleeding and inflammation during tolerization could potentially ease the treatment burden of ITI, data regarding outcomes, annualized bleeding rates, and best dosing regimens of concomitant ITI regimens with emicizumab have yet to be assessed and are currently the object of research.[70] [71]
#
Gene Therapy
Gene therapy has been increasingly investigated during the last two decades, considering that it is the only treatment aimed at definitively curing hemophilia A and B.[72] The adeno-associated virus (AAV) represented the first vector associated with gene transfer in hemophilia animal models and, so far, AAV vectors (mainly AAV5 and AAV8) are the only vectors demonstrated to be able to achieve therapeutic levels of FVIII and FIX in hemophilia patients.[73] [74] A number of AAV vector-mediated gene therapy trials for hemophilia A and B have been conducted in humans so far or are currently ongoing.[48] In the beginning, gene transfer was performed preferentially in hemophilia B because the F9 gene is smaller than the F8 gene, which made it easier to pack the F9 cDNA in AAV vectors. This problem of the larger size of the F8 gene in hemophilia A was solved by using B-domain deleted human FVIII cDNA with a liver-specific promoter.[75] In addition, use of the gain-of-function F9 Padua gene mutation has improved FIX expression levels from these vectors, thus enabling lower vector doses to be used.[76] One hemophilia B trial found that F9 expression was sustained for 8 years after transgene delivery,[77] whereas F9 transgene expression in an earlier AAV-based trial had been relatively short-lived, with decreases being documented after 4 to 6 weeks.[78] Regarding gene therapy in hemophilia A, F8 expression in the longest trial has shown a decline over 4 years although still providing clinical benefit.[79] A more recent hemophilia A trial has demonstrated sustained F8 expression for more than 2 years, which suggests that it may be possible to achieve durable F8 transgene expression.[80] Finally, a phase 3 trial of sustained endogenous production of FVIII using the AAV5-hFVIII-SQ (Valoctocogene roxaparvovec) vector recently provided striking clinical results.[81] All in all, although the interim results of such trials are quite promising, several important issues regarding hemophilia gene therapy remain unsolved. First, there is the problem of achieving persistent expression of a non-integrating vector, such as AAV. The cellular immune response against transduced hepatocytes that present AAV capsid peptides may contribute to the observed lack of persistent transgene expression. The optimal immunosuppressive therapy does, however, still need to be determined. Other problems are the variability in the levels of FVIII and FIX expression among subjects participating in clinical trials and the unpredictability of the responses seen in both hemophilia A and B gene therapy trials. Finally, safety concerns regarding integration and a potential risk of malignancy can only be put to rest by long-term follow-up.[20]
#
Conclusion
Although the treatment of hemophilia has been in continuous evolution over the last 30 years, there have been amazing therapeutic advances in the last decade. The introduction of EHL recombinant FVIII and FIX has been a major advance in the management of hemophilia patients, enabling greater personalization of dosing regimens, with the ideal goal of completely avoiding, through higher trough plasma levels of the clotting factor, all spontaneous bleeds (zero bleeding). Although the newer products have been commercially available for less than 10 years, preliminary real-world data on their use suggest that they have a good safety profile, with no particular concerns regarding the risk of more inhibitor development.
Moreover, innovative products are available (e.g., emicizumab) or are in an advanced stage of development. These products are no longer based simply on replacement of the deficient clotting factor, but have new mechanisms of action. Emicizumab, thanks to its long half-life and the more acceptable subcutaneous route of administration has revolutionized hemophilia therapy, particularly in patients with inhibitors.
Finally, gene therapy is the only way to cure hemophilia definitively through the correction of the cause of this inherited bleeding disorder, although a number of critical issues must be resolved before it can become a first-line therapy for severe hemophilia. Nevertheless, the potential of gene therapy to maintain appropriate clotting factor levels and sustained expression over many years, releasing patients from the burden of injections, makes it clearly the most attractive potential therapeutic weapon and certainly the main field of clinical research in hemophilia in the coming decade.
#
#
Conflict of Interest
P.M.M. reports other from Bayer, personal fees from Kedrion, personal fees from Roche, during the conduct of the study.
-
References
- 1 Mannucci PM, Tuddenham EGD. The hemophilias—from royal genes to gene therapy. N Engl J Med 2001; 344 (23) 1773-1779
- 2 Bolton-Maggs PH, Pasi KJ. Haemophilias A and B. Lancet 2003; 361 (9371): 1801-1809
- 3 Peyvandi F, Garagiola I, Young G. The past and future of haemophilia: diagnosis, treatments, and its complications. Lancet 2016; 388 (10040): 187-197
- 4 Franchini M, Mannucci PM. Past, present and future of hemophilia: a narrative review. Orphanet J Rare Dis 2012; 7: 24
- 5 Franchini M, Mannucci PM. Hemophilia A in the third millennium. Blood Rev 2013; 27 (04) 179-184
- 6 Franchini M, Mannucci PM. The history of hemophilia. Semin Thromb Hemost 2014; 40 (05) 571-576
- 7 Schramm W. The history of haemophilia—a short review. Thromb Res 2014; 134 (Suppl. 01) S4-S9
- 8 Rogaev EI, Grigorenko AP, Faskhutdinova G, Kittler EL, Moliaka YK. Genotype analysis identifies the cause of the “royal disease”. Science 2009; 326 (5954): 817
- 9 Mannucci PM. Back to the future: a recent history of haemophilia treatment. Haemophilia 2008; 14 (Suppl. 03) 10-18
- 10 Mannucci PM. Hemophilia: treatment options in the twenty-first century. J Thromb Haemost 2003; 1 (07) 1349-1355
- 11 Pool JG, Shannon AE. Production of high-potency concentrates of antihemophilic globulin in a closed-bag system. N Engl J Med 1965; 273 (27) 1443-1447
- 12 Nilsson IM. Experience with prophylaxis in Sweden. Semin Hematol 1993; 30 (03, Suppl 2): 16-19
- 13 Moreno MM, Cuesta-Barriuso R. A history of prophylaxis in haemophilia. Blood Coagul Fibrinolysis 2019; 30 (02) 55-57
- 14 Mannucci PM. Desmopressin (DDAVP) in the treatment of bleeding disorders: the first twenty years. Haemophilia 2000; 6 (Suppl. 01) 60-67
- 15 Evatt BL. The tragic history of AIDS in the hemophilia population, 1982-1984. J Thromb Haemost 2006; 4 (11) 2295-2301
- 16 Franchini M. The modern treatment of haemophilia: a narrative review. Blood Transfus 2013; 11 (02) 178-182
- 17 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 (06) 535-544
- 18 Gringeri A, Lundin B, von Mackensen S, Mantovani L, Mannucci PM. ESPRIT Study Group. A randomized clinical trial of prophylaxis in children with hemophilia A (the ESPRIT Study). J Thromb Haemost 2011; 9 (04) 700-710
- 19 Franchini M, Mannucci PM. The safety of pharmacologic options for the treatment of persons with hemophilia. Expert Opin Drug Saf 2016; 15 (10) 1391-1400
- 20 Mannucci PM. Hemophilia therapy: the future has begun. Haematologica 2020; 105 (03) 545-553
- 21 Schulte S. Innovative coagulation factors: albumin fusion technology and recombinant single-chain factor VIII. Thromb Res 2013; 131 (Suppl. 02) S2-S6
- 22 Ivens IA, Baumann A, McDonald TA, Humphries TJ, Michaels LA, Mathew P. PEGylated therapeutic proteins for haemophilia treatment: a review for haemophilia caregivers. Haemophilia 2013; 19 (01) 11-20
- 23 Swierczewska M, Lee KC, Lee S. What is the future of PEGylated therapies?. Expert Opin Emerg Drugs 2015; 20 (04) 531-536
- 24 Aledort L, Mannucci PM, Schramm W, Tarantino M. Factor VIII replacement is still the standard of care in haemophilia A. Blood Transfus 2019; 17 (06) 479-486
- 25 Hermans C, Mancuso ME, Nolan B, Pasi KJ. Recombinant factor VIII Fc for the treatment of haemophilia A. Eur J Haematol 2021; 106 (06) 745-761
- 26 Hermans C, Reding MT, Astermark J, Klamroth R, Mancuso ME. Clinical studies of extended-half-life recombinant FVIII products for prophylaxis in adults and children: a critical review from the physician's perspective. Crit Rev Oncol Hematol 2022; 174: 103678
- 27 Shapiro AD, Ragni MV, Valentino LA. et al. Recombinant factor IX-Fc fusion protein (rFIXFc) demonstrates safety and prolonged activity in a phase 1/2a study in hemophilia B patients. Blood 2012; 119 (03) 666-672
- 28 Powell JS, Pasi KJ, Ragni MV. et al; B-LONG Investigators. Phase 3 study of recombinant factor IX Fc fusion protein in hemophilia B. N Engl J Med 2013; 369 (24) 2313-2323
- 29 Nolan B, Klukowska A, Shapiro A. et al. Final results of the PUPs B-LONG study: evaluating safety and efficacy of rFIXFc in previously untreated patients with hemophilia B. Blood Adv 2021; 5 (13) 2732-2739
- 30 Collins PW, Young G, Knobe K. et al; paradigm 2 Investigators. Recombinant long-acting glycoPEGylated factor IX in hemophilia B: a multinational randomized phase 3 trial. Blood 2014; 124 (26) 3880-3886
- 31 Chan AK, Alamelu J, Barnes C. et al. Nonacog beta pegol (N9-GP) in hemophilia B: first report on safety and efficacy in previously untreated and minimally treated patients. Res Pract Thromb Haemost 2020; 4 (07) 1101-1113
- 32 Santagostino E, Martinowitz U, Lissitchkov T. et al; PROLONG-9FP Investigators Study Group. Long-acting recombinant coagulation factor IX albumin fusion protein (rIX-FP) in hemophilia B: results of a phase 3 trial. Blood 2016; 127 (14) 1761-1769
- 33 Mahlangu J, Powell JS, Ragni MV. et al; A-LONG Investigators. Phase 3 study of recombinant factor VIII Fc fusion protein in severe hemophilia A. Blood 2014; 123 (03) 317-325
- 34 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 (09) 1078-1085
- 35 Giangrande P, Andreeva T, Chowdary P. et al; Pathfinder™2 Investigators. Clinical evaluation of glycoPEGylated recombinant FVIII: efficacy and safety in severe haemophilia A. Thromb Haemost 2017; 117 (02) 252-261
- 36 Mullins ES, Stasyshyn O, Alvarez-Román MT. et al. Extended half-life pegylated, full-length recombinant factor VIII for prophylaxis in children with severe haemophilia A. Haemophilia 2017; 23 (02) 238-246
- 37 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 (03) 411-419
- 38 Nolan B, Mahlangu J, Pabinger I. et al. Recombinant factor VIII Fc fusion protein for the treatment of severe haemophilia A: final results from the ASPIRE extension study. Haemophilia 2020; 26 (03) 494-502
- 39 Chowdary P, Carcao M, Holme PA. et al. Fixed doses of N8-GP prophylaxis maintain moderate-to-mild factor VIII levels in the majority of patients with severe hemophilia A. Res Pract Thromb Haemost 2019; 3 (03) 542-554
- 40 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 (09) 1705-1713
- 41 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
- 42 Königs C, Ozelo MC, Dunn A. et al. First study of extended half-life rFVIIIFc in previously untreated patients with hemophilia A: PUPs A-LONG final results. Blood 2022; 139 (26) 3699-3707
- 43 Pipe SW, Montgomery RR, Pratt KP, Lenting PJ, Lillicrap D. Life in the shadow of a dominant partner: the FVIII-VWF association and its clinical implications for hemophilia A. Blood 2016; 128 (16) 2007-2016
- 44 Konkle BA, Shapiro AD, Quon DV. et al. BIVV001 fusion protein as factor VIII replacement therapy for hemophilia A. N Engl J Med 2020; 383 (11) 1018-1027
- 45 Mancuso ME, Santagostino E. Outcome of clinical trials with new extended half-life FVIII/IX concentrates. J Clin Med 2017; 6 (04) 39
- 46 Gruppo R, López-Fernández MF, Wynn TT, Engl W, Sharkhawy M, Tangada S. 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 (05) 773-781
- 47 Santagostino E, Negrier C, Klamroth R. et al. Safety and pharmacokinetics of a novel recombinant fusion protein linking coagulation factor IX with albumin (rIX-FP) in hemophilia B patients. Blood 2012; 120 (12) 2405-2411
- 48 Ozelo MC, Yamaguti-Hayakawa GG. Impact of novel hemophilia therapies around the world. Res Pract Thromb Haemost 2022; 6 (03) e12695
- 49 Franchini M, Mannucci PM. Inhibitors of propagation of coagulation (factors VIII, IX and XI): a review of current therapeutic practice. Br J Clin Pharmacol 2011; 72 (04) 553-562
- 50 Astermark J, Santagostino E, Keith Hoots W. Clinical issues in inhibitors. Haemophilia 2010; 16 (Suppl. 05) 54-60
- 51 Male C, Andersson NG, Rafowicz A. et al. Inhibitor incidence in an unselected cohort of previously untreated patients with severe haemophilia B: a PedNet study. Haematologica 2021; 106 (01) 123-129
- 52 Meeks SL, Batsuli G. Hemophilia and inhibitors: current treatment options and potential new therapeutic approaches. Hematology (Am Soc Hematol Educ Program) 2016; 2016 (01) 657-662
- 53 Peyvandi F, Mannucci PM, Garagiola I. et al. A randomized trial of factor VIII and neutralizing antibodies in hemophilia A. N Engl J Med 2016; 374 (21) 2054-2064
- 54 Astermark J, Donfield SM, DiMichele DM. et al; FENOC Study Group. A randomized comparison of bypassing agents in hemophilia complicated by an inhibitor: the FEIBA NovoSeven Comparative (FENOC) Study. Blood 2007; 109 (02) 546-551
- 55 Knight C, Danø AM, Kennedy-Martin T. Systematic review of efficacy of rFVIIa and aPCC treatment for hemophilia patients with inhibitors. Adv Ther 2009; 26 (01) 68-88
- 56 Treur MJ, McCracken F, Heeg B. et al. Efficacy of recombinant activated factor VII vs. activated prothrombin complex concentrate for patients suffering from haemophilia complicated with inhibitors: a Bayesian meta-regression. Haemophilia 2009; 15 (02) 420-436
- 57 Franchini M, Coppola A, Tagliaferri A, Lippi G. FEIBA versus NovoSeven in hemophilia patients with inhibitors. Semin Thromb Hemost 2013; 39 (07) 772-778
- 58 Mannucci PM, Mancuso ME, Santagostino E, Franchini M. Innovative pharmacological therapies for the hemophilias not based on deficient factor replacement. Semin Thromb Hemost 2016; 42 (05) 526-532
- 59 Franchini M, Mannucci PM. Non-factor replacement therapy for haemophilia: a current update. Blood Transfus 2018; 16 (05) 457-461
- 60 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 (10) 1570-1574
- 61 Uchida N, Sambe T, Yoneyama K. et al. A first-in-human phase 1 study of ACE910, a novel factor VIII-mimetic bispecific antibody, in healthy subjects. Blood 2016; 127 (13) 1633-1641
- 62 Shima M, Hanabusa H, Taki M. et al. Factor VIII-mimetic function of humanized bispecific antibody in hemophilia A. N Engl J Med 2016; 374 (21) 2044-2053
- 63 Oldenburg J, Mahlangu JN, Kim B. et al. Emicizumab prophylaxis in hemophilia A with inhibitors. N Engl J Med 2017; 377 (09) 809-818
- 64 Callaghan MU, Negrier C, Paz-Priel I. et al. Long-term outcomes with emicizumab prophylaxis for hemophilia A with or without FVIII inhibitors from the HAVEN 1-4 studies. Blood 2021; 137 (16) 2231-2242
- 65 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 (09) 811-822
- 66 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 (06) e295-e305
- 67 Samuelson Bannow B, Recht M, Négrier C. et al. Factor VIII: Long-established role in haemophilia A and emerging evidence beyond haemostasis. Blood Rev 2019; 35: 43-50
- 68 Coppola A, Castaman G, Santoro RC. et al. ad hoc Working Group. Management of patients with severe haemophilia A without inhibitors on prophylaxis with emicizumab: AICE recommendations with focus on emergency in collaboration with SIBioC, SIMEU, SIMEUP, SIPMeL and SISET. Haemophilia 2020; 26: 937-945
- 69 Castaman G, Santoro C, Coppola A. et al; ad hoc Working Group. Emergency management in patients with haemophilia A and inhibitors on prophylaxis with emicizumab: AICE practical guidance in collaboration with SIBioC, SIMEU, SIMEUP, SIPMeL and SISET. Blood Transfus 2020; 18 (02) 143-151
- 70 Le Quellec S, Negrier C. Emicizumab should be prescribed independent of immune tolerance induction. Blood Adv 2018; 2 (20) 2783-2786
- 71 Santagostino E, Young G, Escuriola Ettingshausen C, Jimenez-Yuste V, Carcao M. Inhibitors: a need for eradication?. Acta Haematol 2019; 141 (03) 151-155
- 72 Jair Lara-Navarro I, Rebeca Jaloma-Cruz A. Current therapies in hemophilia: from plasma-derived factor modalities to CRISPR/Cas alternatives. Tohoku J Exp Med 2022; 256 (03) 197-207
- 73 Wang L, Takabe K, Bidlingmaier SM, Ill CR, Verma IM. Sustained correction of bleeding disorder in hemophilia B mice by gene therapy. Proc Natl Acad Sci U S A 1999; 96 (07) 3906-3910
- 74 Chao H, Mao L, Bruce AT, Walsh CE. Sustained expression of human factor VIII in mice using a parvovirus-based vector. Blood 2000; 95 (05) 1594-1599
- 75 Ward NJ, Buckley SM, Waddington SN. et al. Codon optimization of human factor VIII cDNAs leads to high-level expression. Blood 2011; 117 (03) 798-807
- 76 Samelson-Jones BJ, Finn JD, Raffini LJ. et al. Evolutionary insights into coagulation factor IX Padua and other high-specific-activity variants. Blood Adv 2021; 5 (05) 1324-1332
- 77 Nathwani AC, Reiss U, Tuddenham E. et al. Adeno-associated mediated gene transfer for hemophilia B: 8 year follow up and impact of removing “empty viral particles” on safety and efficacy of gene transfer. Blood 2018; 132 (Suppl. 01) 491
- 78 Manno CS, Pierce GF, Arruda VR. et al. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med 2006; 12 (03) 342-347
- 79 Pasi KJ, Rangarajan S, Mitchell N. et al. Multiyear follow-up of AAV5-hFVIII-SQ gene therapy for hemophilia A. N Engl J Med 2020; 382 (01) 29-40
- 80 George LA, Monahan PE, Eyster ME. et al. Multiyear factor VIII expression after AAV gene transfer for hemophilia A. N Engl J Med 2021; 385 (21) 1961-1973
- 81 Ozelo MC, Mahlangu J, Pasi KJ. et al; GENEr8-1 Trial Group. Valoctocogene roxaparvovec gene therapy for hemophilia A. N Engl J Med 2022; 386 (11) 1013-1025
- 82 Miesbach W, Baghaei F, Boban A. et al. Gene therapy of hemophilia: Hub centres should be haemophilia centres: a joint publication of EAHAD and EHC. Haemophilia 2022; 28 (03) e86-e88
Address for correspondence
Publication History
Article published online:
15 September 2022
© 2022. Thieme. All rights reserved.
Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA
-
References
- 1 Mannucci PM, Tuddenham EGD. The hemophilias—from royal genes to gene therapy. N Engl J Med 2001; 344 (23) 1773-1779
- 2 Bolton-Maggs PH, Pasi KJ. Haemophilias A and B. Lancet 2003; 361 (9371): 1801-1809
- 3 Peyvandi F, Garagiola I, Young G. The past and future of haemophilia: diagnosis, treatments, and its complications. Lancet 2016; 388 (10040): 187-197
- 4 Franchini M, Mannucci PM. Past, present and future of hemophilia: a narrative review. Orphanet J Rare Dis 2012; 7: 24
- 5 Franchini M, Mannucci PM. Hemophilia A in the third millennium. Blood Rev 2013; 27 (04) 179-184
- 6 Franchini M, Mannucci PM. The history of hemophilia. Semin Thromb Hemost 2014; 40 (05) 571-576
- 7 Schramm W. The history of haemophilia—a short review. Thromb Res 2014; 134 (Suppl. 01) S4-S9
- 8 Rogaev EI, Grigorenko AP, Faskhutdinova G, Kittler EL, Moliaka YK. Genotype analysis identifies the cause of the “royal disease”. Science 2009; 326 (5954): 817
- 9 Mannucci PM. Back to the future: a recent history of haemophilia treatment. Haemophilia 2008; 14 (Suppl. 03) 10-18
- 10 Mannucci PM. Hemophilia: treatment options in the twenty-first century. J Thromb Haemost 2003; 1 (07) 1349-1355
- 11 Pool JG, Shannon AE. Production of high-potency concentrates of antihemophilic globulin in a closed-bag system. N Engl J Med 1965; 273 (27) 1443-1447
- 12 Nilsson IM. Experience with prophylaxis in Sweden. Semin Hematol 1993; 30 (03, Suppl 2): 16-19
- 13 Moreno MM, Cuesta-Barriuso R. A history of prophylaxis in haemophilia. Blood Coagul Fibrinolysis 2019; 30 (02) 55-57
- 14 Mannucci PM. Desmopressin (DDAVP) in the treatment of bleeding disorders: the first twenty years. Haemophilia 2000; 6 (Suppl. 01) 60-67
- 15 Evatt BL. The tragic history of AIDS in the hemophilia population, 1982-1984. J Thromb Haemost 2006; 4 (11) 2295-2301
- 16 Franchini M. The modern treatment of haemophilia: a narrative review. Blood Transfus 2013; 11 (02) 178-182
- 17 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 (06) 535-544
- 18 Gringeri A, Lundin B, von Mackensen S, Mantovani L, Mannucci PM. ESPRIT Study Group. A randomized clinical trial of prophylaxis in children with hemophilia A (the ESPRIT Study). J Thromb Haemost 2011; 9 (04) 700-710
- 19 Franchini M, Mannucci PM. The safety of pharmacologic options for the treatment of persons with hemophilia. Expert Opin Drug Saf 2016; 15 (10) 1391-1400
- 20 Mannucci PM. Hemophilia therapy: the future has begun. Haematologica 2020; 105 (03) 545-553
- 21 Schulte S. Innovative coagulation factors: albumin fusion technology and recombinant single-chain factor VIII. Thromb Res 2013; 131 (Suppl. 02) S2-S6
- 22 Ivens IA, Baumann A, McDonald TA, Humphries TJ, Michaels LA, Mathew P. PEGylated therapeutic proteins for haemophilia treatment: a review for haemophilia caregivers. Haemophilia 2013; 19 (01) 11-20
- 23 Swierczewska M, Lee KC, Lee S. What is the future of PEGylated therapies?. Expert Opin Emerg Drugs 2015; 20 (04) 531-536
- 24 Aledort L, Mannucci PM, Schramm W, Tarantino M. Factor VIII replacement is still the standard of care in haemophilia A. Blood Transfus 2019; 17 (06) 479-486
- 25 Hermans C, Mancuso ME, Nolan B, Pasi KJ. Recombinant factor VIII Fc for the treatment of haemophilia A. Eur J Haematol 2021; 106 (06) 745-761
- 26 Hermans C, Reding MT, Astermark J, Klamroth R, Mancuso ME. Clinical studies of extended-half-life recombinant FVIII products for prophylaxis in adults and children: a critical review from the physician's perspective. Crit Rev Oncol Hematol 2022; 174: 103678
- 27 Shapiro AD, Ragni MV, Valentino LA. et al. Recombinant factor IX-Fc fusion protein (rFIXFc) demonstrates safety and prolonged activity in a phase 1/2a study in hemophilia B patients. Blood 2012; 119 (03) 666-672
- 28 Powell JS, Pasi KJ, Ragni MV. et al; B-LONG Investigators. Phase 3 study of recombinant factor IX Fc fusion protein in hemophilia B. N Engl J Med 2013; 369 (24) 2313-2323
- 29 Nolan B, Klukowska A, Shapiro A. et al. Final results of the PUPs B-LONG study: evaluating safety and efficacy of rFIXFc in previously untreated patients with hemophilia B. Blood Adv 2021; 5 (13) 2732-2739
- 30 Collins PW, Young G, Knobe K. et al; paradigm 2 Investigators. Recombinant long-acting glycoPEGylated factor IX in hemophilia B: a multinational randomized phase 3 trial. Blood 2014; 124 (26) 3880-3886
- 31 Chan AK, Alamelu J, Barnes C. et al. Nonacog beta pegol (N9-GP) in hemophilia B: first report on safety and efficacy in previously untreated and minimally treated patients. Res Pract Thromb Haemost 2020; 4 (07) 1101-1113
- 32 Santagostino E, Martinowitz U, Lissitchkov T. et al; PROLONG-9FP Investigators Study Group. Long-acting recombinant coagulation factor IX albumin fusion protein (rIX-FP) in hemophilia B: results of a phase 3 trial. Blood 2016; 127 (14) 1761-1769
- 33 Mahlangu J, Powell JS, Ragni MV. et al; A-LONG Investigators. Phase 3 study of recombinant factor VIII Fc fusion protein in severe hemophilia A. Blood 2014; 123 (03) 317-325
- 34 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 (09) 1078-1085
- 35 Giangrande P, Andreeva T, Chowdary P. et al; Pathfinder™2 Investigators. Clinical evaluation of glycoPEGylated recombinant FVIII: efficacy and safety in severe haemophilia A. Thromb Haemost 2017; 117 (02) 252-261
- 36 Mullins ES, Stasyshyn O, Alvarez-Román MT. et al. Extended half-life pegylated, full-length recombinant factor VIII for prophylaxis in children with severe haemophilia A. Haemophilia 2017; 23 (02) 238-246
- 37 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 (03) 411-419
- 38 Nolan B, Mahlangu J, Pabinger I. et al. Recombinant factor VIII Fc fusion protein for the treatment of severe haemophilia A: final results from the ASPIRE extension study. Haemophilia 2020; 26 (03) 494-502
- 39 Chowdary P, Carcao M, Holme PA. et al. Fixed doses of N8-GP prophylaxis maintain moderate-to-mild factor VIII levels in the majority of patients with severe hemophilia A. Res Pract Thromb Haemost 2019; 3 (03) 542-554
- 40 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 (09) 1705-1713
- 41 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
- 42 Königs C, Ozelo MC, Dunn A. et al. First study of extended half-life rFVIIIFc in previously untreated patients with hemophilia A: PUPs A-LONG final results. Blood 2022; 139 (26) 3699-3707
- 43 Pipe SW, Montgomery RR, Pratt KP, Lenting PJ, Lillicrap D. Life in the shadow of a dominant partner: the FVIII-VWF association and its clinical implications for hemophilia A. Blood 2016; 128 (16) 2007-2016
- 44 Konkle BA, Shapiro AD, Quon DV. et al. BIVV001 fusion protein as factor VIII replacement therapy for hemophilia A. N Engl J Med 2020; 383 (11) 1018-1027
- 45 Mancuso ME, Santagostino E. Outcome of clinical trials with new extended half-life FVIII/IX concentrates. J Clin Med 2017; 6 (04) 39
- 46 Gruppo R, López-Fernández MF, Wynn TT, Engl W, Sharkhawy M, Tangada S. 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 (05) 773-781
- 47 Santagostino E, Negrier C, Klamroth R. et al. Safety and pharmacokinetics of a novel recombinant fusion protein linking coagulation factor IX with albumin (rIX-FP) in hemophilia B patients. Blood 2012; 120 (12) 2405-2411
- 48 Ozelo MC, Yamaguti-Hayakawa GG. Impact of novel hemophilia therapies around the world. Res Pract Thromb Haemost 2022; 6 (03) e12695
- 49 Franchini M, Mannucci PM. Inhibitors of propagation of coagulation (factors VIII, IX and XI): a review of current therapeutic practice. Br J Clin Pharmacol 2011; 72 (04) 553-562
- 50 Astermark J, Santagostino E, Keith Hoots W. Clinical issues in inhibitors. Haemophilia 2010; 16 (Suppl. 05) 54-60
- 51 Male C, Andersson NG, Rafowicz A. et al. Inhibitor incidence in an unselected cohort of previously untreated patients with severe haemophilia B: a PedNet study. Haematologica 2021; 106 (01) 123-129
- 52 Meeks SL, Batsuli G. Hemophilia and inhibitors: current treatment options and potential new therapeutic approaches. Hematology (Am Soc Hematol Educ Program) 2016; 2016 (01) 657-662
- 53 Peyvandi F, Mannucci PM, Garagiola I. et al. A randomized trial of factor VIII and neutralizing antibodies in hemophilia A. N Engl J Med 2016; 374 (21) 2054-2064
- 54 Astermark J, Donfield SM, DiMichele DM. et al; FENOC Study Group. A randomized comparison of bypassing agents in hemophilia complicated by an inhibitor: the FEIBA NovoSeven Comparative (FENOC) Study. Blood 2007; 109 (02) 546-551
- 55 Knight C, Danø AM, Kennedy-Martin T. Systematic review of efficacy of rFVIIa and aPCC treatment for hemophilia patients with inhibitors. Adv Ther 2009; 26 (01) 68-88
- 56 Treur MJ, McCracken F, Heeg B. et al. Efficacy of recombinant activated factor VII vs. activated prothrombin complex concentrate for patients suffering from haemophilia complicated with inhibitors: a Bayesian meta-regression. Haemophilia 2009; 15 (02) 420-436
- 57 Franchini M, Coppola A, Tagliaferri A, Lippi G. FEIBA versus NovoSeven in hemophilia patients with inhibitors. Semin Thromb Hemost 2013; 39 (07) 772-778
- 58 Mannucci PM, Mancuso ME, Santagostino E, Franchini M. Innovative pharmacological therapies for the hemophilias not based on deficient factor replacement. Semin Thromb Hemost 2016; 42 (05) 526-532
- 59 Franchini M, Mannucci PM. Non-factor replacement therapy for haemophilia: a current update. Blood Transfus 2018; 16 (05) 457-461
- 60 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 (10) 1570-1574
- 61 Uchida N, Sambe T, Yoneyama K. et al. A first-in-human phase 1 study of ACE910, a novel factor VIII-mimetic bispecific antibody, in healthy subjects. Blood 2016; 127 (13) 1633-1641
- 62 Shima M, Hanabusa H, Taki M. et al. Factor VIII-mimetic function of humanized bispecific antibody in hemophilia A. N Engl J Med 2016; 374 (21) 2044-2053
- 63 Oldenburg J, Mahlangu JN, Kim B. et al. Emicizumab prophylaxis in hemophilia A with inhibitors. N Engl J Med 2017; 377 (09) 809-818
- 64 Callaghan MU, Negrier C, Paz-Priel I. et al. Long-term outcomes with emicizumab prophylaxis for hemophilia A with or without FVIII inhibitors from the HAVEN 1-4 studies. Blood 2021; 137 (16) 2231-2242
- 65 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 (09) 811-822
- 66 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 (06) e295-e305
- 67 Samuelson Bannow B, Recht M, Négrier C. et al. Factor VIII: Long-established role in haemophilia A and emerging evidence beyond haemostasis. Blood Rev 2019; 35: 43-50
- 68 Coppola A, Castaman G, Santoro RC. et al. ad hoc Working Group. Management of patients with severe haemophilia A without inhibitors on prophylaxis with emicizumab: AICE recommendations with focus on emergency in collaboration with SIBioC, SIMEU, SIMEUP, SIPMeL and SISET. Haemophilia 2020; 26: 937-945
- 69 Castaman G, Santoro C, Coppola A. et al; ad hoc Working Group. Emergency management in patients with haemophilia A and inhibitors on prophylaxis with emicizumab: AICE practical guidance in collaboration with SIBioC, SIMEU, SIMEUP, SIPMeL and SISET. Blood Transfus 2020; 18 (02) 143-151
- 70 Le Quellec S, Negrier C. Emicizumab should be prescribed independent of immune tolerance induction. Blood Adv 2018; 2 (20) 2783-2786
- 71 Santagostino E, Young G, Escuriola Ettingshausen C, Jimenez-Yuste V, Carcao M. Inhibitors: a need for eradication?. Acta Haematol 2019; 141 (03) 151-155
- 72 Jair Lara-Navarro I, Rebeca Jaloma-Cruz A. Current therapies in hemophilia: from plasma-derived factor modalities to CRISPR/Cas alternatives. Tohoku J Exp Med 2022; 256 (03) 197-207
- 73 Wang L, Takabe K, Bidlingmaier SM, Ill CR, Verma IM. Sustained correction of bleeding disorder in hemophilia B mice by gene therapy. Proc Natl Acad Sci U S A 1999; 96 (07) 3906-3910
- 74 Chao H, Mao L, Bruce AT, Walsh CE. Sustained expression of human factor VIII in mice using a parvovirus-based vector. Blood 2000; 95 (05) 1594-1599
- 75 Ward NJ, Buckley SM, Waddington SN. et al. Codon optimization of human factor VIII cDNAs leads to high-level expression. Blood 2011; 117 (03) 798-807
- 76 Samelson-Jones BJ, Finn JD, Raffini LJ. et al. Evolutionary insights into coagulation factor IX Padua and other high-specific-activity variants. Blood Adv 2021; 5 (05) 1324-1332
- 77 Nathwani AC, Reiss U, Tuddenham E. et al. Adeno-associated mediated gene transfer for hemophilia B: 8 year follow up and impact of removing “empty viral particles” on safety and efficacy of gene transfer. Blood 2018; 132 (Suppl. 01) 491
- 78 Manno CS, Pierce GF, Arruda VR. et al. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med 2006; 12 (03) 342-347
- 79 Pasi KJ, Rangarajan S, Mitchell N. et al. Multiyear follow-up of AAV5-hFVIII-SQ gene therapy for hemophilia A. N Engl J Med 2020; 382 (01) 29-40
- 80 George LA, Monahan PE, Eyster ME. et al. Multiyear factor VIII expression after AAV gene transfer for hemophilia A. N Engl J Med 2021; 385 (21) 1961-1973
- 81 Ozelo MC, Mahlangu J, Pasi KJ. et al; GENEr8-1 Trial Group. Valoctocogene roxaparvovec gene therapy for hemophilia A. N Engl J Med 2022; 386 (11) 1013-1025
- 82 Miesbach W, Baghaei F, Boban A. et al. Gene therapy of hemophilia: Hub centres should be haemophilia centres: a joint publication of EAHAD and EHC. Haemophilia 2022; 28 (03) e86-e88