3,266
Views
1
CrossRef citations to date
0
Altmetric
Review

New directions to develop therapies for people with hemophilia

ORCID Icon, , &
Pages 417-433 | Received 09 Nov 2022, Accepted 21 Feb 2023, Published online: 08 Mar 2023

ABSTRACT

Introduction

The past few decades have seen a tremendous advancement in the management of hemophilia. Whether it is improved methods to attenuate critical viruses, recombinant bioengineering with decreased immunogenicity, extended half-life replacement therapies to mitigate the burden of repeated infusion treatments, novel nonreplacement products to avoid the drawback of inhibitor development with its attractive subcutaneous administration and then the introduction of gene therapy, the management has trodden a long way.

Areas covered

This expert review describes the progress in the treatment of hemophilia over the years. We discuss, in detail, the past and current therapies, their benefits, drawbacks, along with relevant studies leading to approval, efficacy and safety profile, ongoing trials, and future prospects.

Expert opinion

The technological advances in the treatment of hemophilia with convenient modes of administration and innovative modalities offer a chance for a normal existence of the patients living with this disease. However, it is imperative for clinicians to be aware of the potential adverse effects and the need for further studies to establish causality or chance association of these events with novel agents. Thus, it is crucial for clinicians to engage patients and their families in informed decision-making and tailor individual concerns and necessities.

1. Introduction

Hemophilia A (HA) and B (HB) are X linked recessively inherited monogenic bleeding disorders caused by quantitative or qualitative abnormalities of coagulation factor VIII (FVIII) and factor IX (FIX), respectively [Citation1]. The first inscriptional account of hemophilia dates back to the 2nd century AD described in the Talmud, Jewish rabbinical writings that mentions to avoid circumcision in children if two of their brothers had previously died due to hemorrhage following the procedure [Citation2]. The modern-day accounts materialized only in the 19th century suggests that the only available treatment during that time was whole blood transfusion. Subsequently, in the middle of the 20th century, the discovery of “antihemophilic globulin” lined the way to the invention of cryoprecipitate and then of FVIII and FIX concentrates [Citation3]. Sadly, in 1980s, calamitous infection with human immunodeficiency virus and hepatitis C virus was reported in hemophiliacs due to contamination of factor concentrate supply thus necessitating development of improved methods to attenuate viruses. Dry-heat, pasteurization, solvent-detergent treatment, immunoaffinity purification, and nanofiltration are the main contrivances used to attenuate this contamination and prevent transmission-related infection [Citation4].

The availability of these factor replacement products has drastically diminished the morbidity and mortality associated with hemophilia and remains the backbone of hemophilia treatment. However, the development of neutralizing alloantibodies i.e. inhibitors, is the most despairing and problematic treatment-related complication that affects at least one-third of individuals with severe hemophilia [Citation5]. Inhibitor formation has an estimated incidence of 30% in severe HA [Citation6] and 9.3% in severe HB [Citation7] The fundamental method for inhibitor eradication is immune tolerance induction which is a technique to tolerize the individual’s immune system to the clotting factor and reduce antibody production via the administration of repeated doses of the factor [Citation8]. This method is intensive, costly and requires frequent and long-term administration of factor concentrates and is associated with significant complications including anaphylactic reactions or complications related to nephrotic syndrome, the latter evident in HB [Citation9]. ITI protocols to eliminate inhibitors are not effective in all patients, hence new experimental therapies are being developed. One such investigational therapy is an oral tolerance protocol based on transgenic lettuce plants that suppressed inhibitor formation in hemophilic mice and dogs [Citation10]. Furthermore, studies are ongoing to determine factors affecting the inhibitor development in hemophiliacs, one such study is a prospective multicenter longitudinal cohort study the “Hemophilia Inhibitor Previously Untreated Patients (PUP) Study” that revealed distinct antibody signatures prior to FVIII inhibitor development [Citation11].

Presently, more effective and less cumbersome treatments have been intended to overcome these unmet needs of current therapeutic strategies, such as non-replacement factor therapies that are effective regardless of the presence of inhibitors. These innovative therapies are bypassing agents that seek to rebalance the hemostatic defect by targeting the natural anticoagulant pathways that regulate hemostasis such as activated prothrombin complex concentrates (aPCC), recombinant activated factor VII [Citation12] and plasma-derived FVIIa/FX complex [Citation13]. Emicizumab-kxwh, that has been licensed for routine use in patients with HA, is a bispecific monoclonal antibody that bridges activated FIX to factor X, mimicking the function of activated FVIII, thereby increasing thrombin formation [Citation14]. Another way besides enhancing coagulation is to inhibit anticoagulant pathways by the use of fitusiran, an antisense oligonucleotide that ultimately decreases transcription and translation of native antithrombin RNA [Citation15], and marstacimab/concizumab that are monoclonal antibodies against tissue factor pathway inhibitor (TFPI) [Citation16,Citation17]. Although studies have reported thrombotic complications with emicizumab, it has been widely used as prophylaxis. Fitusiran remains in development, its clinical trial progress interrupted by unexpected thrombotic complications in a few study subjects [Citation16,Citation18]. Thus, even though the evidence for efficacy of the “non-factor therapies is reassuring, their safety profiles have raised concern, hence caution is necessary when prescribing or encouraging participation in clinical trials.

Another shortcoming of these factor replacement agents is that standard half-life (SHL) coagulation products for prophylaxis typically need 2–4 injections per week. Newer techniques with Fc-fusion technology and PEGylation has led to the development of extended half-life (EHLs) FVIII [Citation19] and FIX that allow longer intervals between dosing schedules [Citation20]. Subcutaneous (SQ) and oral administration that is currently underway would also streamline prophylaxis, improve compliance by eliminating the need of intravenous (IV) access, reducing dosage frequency, and extending protection from bleeding. For over three decades, researchers have ardently pursued efforts to potentially cure hemophilia through the development of gene therapy. Gene therapy includes introduction of either a wild-type factor gene or a modified gene with enhanced properties such as B-domain deletion to facilitate vector loading (FVIII), or using a gain-of-function gene variant to enhance gene product activity (FIX Padua). Hepatocytes are an appropriate target cell for transduction since liver is the site of synthesis of most clotting factors. Diverse technologies have been used to introduce the F8 or F9 gene, either using ex vivo genetic modification followed by implantation of the modified cells or by means of a vector carrying a construct that introduces the normal gene or vectors that produce other coagulation factors such as factor VIIa that can be used in hemophiliacs with immune tolerance [Citation21,Citation22] Autologous bone marrow cells derived mesenchymal stromal cells transduced with a lentiviral vector also showed promising results with elevated FVIII levels [Citation23].

In this article, we will critically analyze the current therapies and highlight the noteworthy advancement in the existing therapeutic resources for the management of patients with HA and HB.

2. Current therapies

Factor VIII or IX replacement, nevertheless, is broadly used with years of undisputable data supported by clinical and real-world experience to treat and prevent bleeds in patients with hemophilia and to prevent long-term complications such as arthropathy. The hallmark longitudinal study by Nilsson set the footing to establish primary prophylaxis to prevent orthopedic complications of repeated bleeds [Citation24]. Primary prophylaxis starts at young age [≤2 years] and is long term, before joint disease develops, whereas secondary prophylaxis begins after the onset of joint disease [Citation25,Citation26].

In 1992, the first commercially available recombinant factor VIII product became available for HA and then subsequently in 1997 for HB [Citation27]. Since then, a plethora of plasma-derived and recombinant products have been developed. The existing standard products have relatively short half-lives (8–12 h for FVIII and 18–24 h for FIX), often necessitating treatments 2–3 times per week [Citation28].

Modern advances, such as EHL products and pharmacokinetic-guided dosing, provide new opportunities to tailor regimes for each individual patient. There are two key techniques of extending the half-life of replacement products. The first is the coagulation factor fusion to proteins like the Fc portion of IgG1 or albumin which extends biological activity by recycling via the neonatal Fc receptor [Citation29]. The second method is the binding with chemicals such as polyethylene glycol (PEG) that acts by slowing their degradation and renal elimination [Citation30].

Despite EHL therapy, hemophilia patients with FVIII inhibitors remain poor candidates for prophylaxis [Citation31] and often require bypassing agents like recombinant human factor VII and aPCC for acute bleeding and preoperatively.

[Citation31] For prophylaxis, non-replacement agents have been studied that act by either mimicking the coagulant activity of FVIII or “rebalancing agents” by increasing defective thrombin formation through the inhibition of the naturally occurring anticoagulants (antithrombin, TFPI, and activated protein C) [Citation32].

To date, only emicizumab that mimics the coagulant activity FVIII activity has been licensed and available commercially [Citation33]. Emicizumab is largely being used now for prophylaxis in patients with HA. Based on phase 2 results, the US Food and Drug Administration (FDA) granted concizumab (previously called mAb2021), a monoclonal antibody against TFPI, “Breakthrough Therapy” designation for HB with inhibitor patients [Citation34].

2.1 Plasma derived Factor VIII and IX products

Once carefully screened donor plasma is designated, the factors are then prepared by commercial fractionation which include either chromatography or monoclonal antibody affinity [Citation35]. Pasteurization, solvent/detergent treatment, chemical disruption with sodium thiocyanate, or ultrafiltration are methods of viral inactivation that augment protection against HIV and hepatitis viruses. These products are thus graded based on purity. Higher purity reflects a higher ratio of FVIII to non-factor VIII proteins [Citation35]. These are divided into Intermediate purity, high purity, and Ultrahigh purity. The main adverse events (AEs) noted with FVIII are increased FVIII inhibitors and inflammation at injection site [Citation36]. AEs with FIX products are rare and involve inhibitor development, skin reactions, and hypersensitivity [Citation36]. The various available plasma-derived products have been described in .

Table 1. Depicting Plasma derived products and their characteristics [Citation36]

: depicts Plasma-derived products and their characteristics [Citation36]

2.2 Recombinant Factor VIII and IX products

Recombinant human FVIII are made using modified versions of the human FVIII gene. They are categorized into four generations with the first three based on the decreasing exposure to animal proteins [Citation37]. Additional modifications of the FVIII gene including B domain deletion and single chain products are manufactured to enhance the pharmacokinetic profile of the product. Examples of “B-domain deleted” products include Xyntha, turcotococog alfa (Novoeight), simoctocog alfa (Nuwiq), Eloctate [Citation38–40]. Multiple clinical trials have demonstrated excellent efficacy and safety profiles [Citation41–44]. Recombinant human FIX is genetically engineered by insertion of the human FIX gene into a Chinese hamster ovary cell line. These products have been demonstrated to be safe and effective in the treatment of previously treated patients (PTPs) and PUPs with HB [Citation45–48].

The recombinant products (second generation and above), however have a safety advantage over plasma-derived concentrates as they have no added albumin [Citation49]. Thus, it is advantageous as it reduces the risk of transmitting animal or human infectious agents. The second-generation octocog alfa (Kogenate FS, Refacto) does not contain albumin and have replaced it with a carbohydrate stabilizer, sucrose [Citation50].

The RODIN study concluded that the risk of developing inhibitors among PUPs was similar in those receiving plasma-derived and recombinant products (95% confidence interval [CI], 0.62 to 1.49). The study also concluded that second-generation recombinant products were associated with an increased risk of inhibitor development (adjusted hazard ratio, 1.60; 95% CI, 1.08 to 2.37).

[Citation51]. However, the 2016 Survey of Inhibitors in Plasma-Product Exposed Toddlers (SIPPET) trial remains the only prospective randomized trial to assess the incidence of FVIII inhibitors among patients treated with plasma-derived FVIII containing von Willebrand factor (vWF) or recombinant FVIII. This study, in contrast showed that the cumulative incidence of all inhibitors was higher with recombinant factor VIII therapy in contrast to plasma derived fVIII (44.5% vs. 26.8%) 44.5%. The cumulative incidence of high-titer inhibitors was 18.6% [95% CI, 11.2 to 26.0] and 28.4% [95% CI, 19.6 to 37.2], respectively [Citation52].The main adverse events noted with rFVIII products are increased factor VIII inhibitors, skin reactions, headache, arthralgia, pyrexia, upper respiratory tract infection and nasopharyngitis [Citation36]. AEs reported with rFIX include antibody development and headache [Citation36]. The various available recombinant FVIII and FIX products have been described in .

Table 2. Depicting Recombinant products and their characteristics [Citation36]

: depicts Recombinant products and their characteristics [Citation36]

2.3 EHL rFVIII products

In 2014, the first EHL was licensed for the management and prevention of bleeding and for perioperative management of individuals with HA. It was the recombinant FVIII Fc-fusion protein Efmoroctocog alfa (Eloctate, Sanofi Genzyme), engineered with recombinant, B-domain deleted, human FVIII fused with a monomeric human immunoglobulin (IgG) Fc domain [Citation53]. This recombinant FVIII Fc-fusion protein demonstrated a 1.5–1.7-fold longer plasma half-life compared with a SHL rFVIII [Citation54]. In various phase III studies, it was found to be well tolerated and efficacious for the management of perioperative hemostasias in patients with severe hemophilia. In the A-LONG trial, for individualized prophylaxis, weekly prophylaxis and episodic treatment arms, the median annualized bleeding rate (ABR) was 1.6, 3.6, and 33.6, with no bleeding episodes occurring in 45.3% and 17.4% of patients in either of the prophylactic arms [Citation55]. Although at-risk patients with a history or evidence of FVIII inhibitors, as well as PUPs, were excluded from the pivotal clinical trial, the use of rFVIII-Fc in these PTPs did not cause any detectable FVIII inhibitors [Citation55–58]. Moreover, it has also been used for ITI in patients with severe HA and high-titer inhibitors and revealed variable levels of success [Citation59].

Another product, Rurioctocog alpha pegol (Adynovate, BAX 855, Takeda Pharmaceutical) that was licensed in 2017 with indications for the treatment and prevention of bleeding and perioperative management of HA [Citation60,Citation61]. The mean half-life of Adynovate was 1.3–1.5-fold longer than that of SHL rFVIII (Advate) with no inhibitors developing in the PTP subjects [Citation62]. The PROLONG-ATE trial demonstrated the efficacy, pharmacokinetics, and safety of BAX 855 for prophylaxis of bleeding and perioperative management of patients with severe HA [Citation63] The 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 [Citation63]. No inhibitors or AEs were reported. Another product with similar technology was licensed in 2018 was the Damoctocog alfa pegol (Jivi, BAY 94–9027, Bayer HealthCare) [Citation64,Citation65]. This product established a longer FVIII half-life and higher area under the curve than a SHL rFVIII (rFVIIIFS, KogenateFS) in a phase I trial in patients with severe HA [Citation64,Citation66] . No developed FVIII inhibitors in the PTP subjects. The PROTECT VIII trial demonstrated the safety and efficacy of BAY 94–9027 in the treatment of patients undergoing major surgeries [Citation67]. The trial demonstrated median ABRs of 4.1, 1.9 and 3.9 in the twice-weekly, every 5 days and every 7 days interval groups, respectively [Citation68]. AEs were typical of events occurring in the hemophilia population. Recent preclinical studies demonstrating complete excretion of PEG-60-Mal-Cys in BAY 94–9027 and no indication of irreversible binding to tissues [Citation69]. . Turoctocog alfa pegol (Esperoct; N8-GP, Novo Nordisk) is yet another PEG rVIII product is also a EHL FVIII product [Citation70]. This product also demonstrated a 1.6-fold longer half-life than patients’ previous FVIII products in PTPs with severe HA [Citation71]. N8-GP was also efficacious and well tolerated in the treatment of patients undergoing major surgeries in the pathfinder 5 trial. No inhibitors or AEs were detected. Patients in the prophylaxis arm had a median ABR of 1.95 and 42.6% of them experienced zero bleeds during the study [Citation72]. The various available EHL FVIII products have been described in .

Table 3. Depicting EHL factor products and their characteristics [Citation36]

2.4 EHL rFIX products

Efrenonacog alfa (Alprolix, Bioverativ Therapeutics Inc.) is a recombinant product composed of FIX fused with a monomeric human immunoglobulin Fc domain was approved by the FDA in 2014 for the prophylaxis and treatment of bleeding in adults with HB, in both the routine and perioperative settings and further approved for pediatric use in 2017. The half-life is three- to fivefold compared with unmodified FIX (i.e half-life of 54 to 90 hours) [Citation73,Citation74]. In a study involving 123 PTPs with HB (age ≥12 years) who received once weekly prophylaxis or as needed prophylaxis ABR were significantly lower than those for patients receiving on-demand therapy (3, 1.4, and 17.7, respectively) [Citation75]. In another study involving previous treated boys younger than 12 years who received once-weekly prophylaxis the median ABR was two for all bleeds and zero for joint bleeds [Citation76]. No PTP patient developed an inhibitor, and no safety concerns were identified [Citation76]. It demonstrates a linear response dose vs efficacy up to 150 IU/kg as it saturates the extravascular compartment and no increased risk for thrombosis at 250 IU/kg, thus doses ∼3 times higher than the currently recommended 40 to 50 IU/kg because of large extravascular compartment, efficiently prolong hemostasis without thrombotic risk [Citation77].

Albutrepenonacog alfa (Idelvion, CSL Behring) is a coagulation FIX [Recombinant], recombinant albumin Fusion Protein (rIX-FP), a recombinant DNA-derived coagulation FIX concentrate first approved by the FDA in 2016 to control and prevent bleeding episodes, management of peri-operative bleeding, and as routine prophylaxis to reduce the frequency of bleeding episodes use in children and adults with HB [Citation78]. This is the first coagulation factor–albumin fusion protein product and the second FIX fusion protein product to be licensed in the United States. The product’s approval is based on results from two multi-center clinical trials that demonstrated the safety and efficacy of albutrepenonacog alfa in 90 adult and pediatric patients with HB (age range = 1–61 years) [Citation20,Citation79]. The half-life of this product is approximately 102 hours (approximately 5.6-fold prolongation). Based on the analysis of the 40 subjects treated with this product for weekly prophylaxis, the estimated mean ABR was 0.83, 0.56, and 0.65 in the 14-, 10-, and 7-day prophylaxis regimens, respectively. No patient developed an inhibitor, and no safety concerns were identified [Citation20].

Another product, Nonacog beta pegol (Rebinyn, N9-GP, Novo Nordisk) is a recombinant FIX product produced in Chinese Hamster Ovary cells composed of the activation sequence of the FIX attached to PEG that was licensed in 2017 [Citation80]. This rFIX demonstrated approximately fivefold prolongation with a half-life of approximately 93 hours [Citation81]. The approval was based on a study involving 74 PTPs (aged 13 to 65 years) with HB, ABR for individuals who received once weekly prophylaxis were significantly lower than that for patients receiving on-demand therapy (1.0, 2.9, and 15.6, respectively) [Citation82]. The therapy was well tolerated, and there were no inhibitors during one year of therapy [Citation82]. The various available EHL FIX products have been described in .

: depicts EHL factor products and their characteristics [Citation36]

2.5 Non-replacement therapies

Emicizumab (Hemlibra, previously designated ACE910) is a bispecific recombinant monoclonal antibody, engineered to bind activated FIX (FIXa) and factor X simultaneously, and thereby promotes thrombin formation by mimicking FVIIIa activity regardless of FVIII deficiency and/or the presence of FVIII inhibitors [Citation83]. It was approved by the FDA for individuals with HA with inhibitors in 2017 and for those without inhibitors in 2018. The recommended dose for HA prophylaxis is 3 mg/kg once weekly for 4 weeks initially followed by 1.5 mg/kg once weekly or 3 mg/kg once every 2 weeks or 6 mg/kg once every 4 weeks as maintenance [Citation84]. Advantages of emicizumab include SQ administration and a long plasma half-life allowing administration every week or even every two weeks [Citation85].

The first two key trials were carried out in patients with FVIII inhibitors were the HAVEN 1 and HAVEN 2. HAVEN 1 included 109 individuals ages 12 years or older and HAVEN 2 trial included 85 children aged 2 to 11 [Citation86,Citation87]. The ABR ranged between 0.2 and 2.9 and the rate of zero bleeding events ranged between 63% and 90% in both the trials. Following the promising results obtained in HAVEN 1 and 2, emicizumab has also been evaluated in patients without inhibitors. In the HAVEN 3 trial, 152 individuals ages 12 years or older with severe HA without inhibitor were enrolled. Study participants had much lower ABR than those not on prophylaxis [1.5 and 1.3 vs. 38.2] and much higher rates of zero bleeds (50% and 40% vs. 0%). In the subgroup of 48 patients previously treated with routine FVIII prophylaxis, transition to emicizumab reduced the ABR by 68% [Citation88] .

Additionally, ABR and the zero bleeding rates appeared to be better than those obtained with the EHL FVIII products, however, there are no head on comparison studies between replacement and non-replacement products. A published matching adjusted indirect comparison for efficacy of rFVIIIFc (in A-LONG trial) versus emicizumab (in HAVEN trial) for mean ABR showed that there were no significant differences between either (with emicizumab administered q1, q2 or q4 weeks). rFVIIIFc has significantly higher proportion of patients with zero bleeds as compared to emicizumab administered 4 weekly but similar to those administered weekly and biweekly [Citation89] . Thus, the difference between either is based on safety profile that is yet to be compared.

One of the complications that was noted in the HAVEN 1 was that a few patients developed thromboses and thrombotic microangiopathies when patients with inhibitor patients had a bleeding episode and were concomitantly treated with large and frequent doses of aPCC [Citation86]. 13 deaths (including in 11 patients with inhibitors) have now been reported since the original clinical trial was published [Citation90]. A post hoc analysis has shown concomitant use of emicizumab and rFVIIa as safe with no reported thrombotic events [Citation91].

Other potential disadvantages may emerge in cases in which FVIII replacement is required to prevent or treat breakthrough bleeds, because the delayed FVIII inhibitors may develop in high-risk circumstances, such as at the time of major trauma or surgery. Another unknown fact is that whether, it provides the same functional benefits of the conventional coagulation factor, such as the long-term preservation of joint health and wound healing [Citation92]. Also, the differences between emicizumab and FVIII also compromises the capacity to interpret FVIII bioequivalence.

3. Future directions and products in development

The current therapies have numerous shortcomings. In hemophilia patients without inhibitors, the reduction in the frequency of IV infusions by means of EHL therapy was still based on the need for a venous access [Citation32]. Hence, newer agents with SQ and oral delivery are underway with recognizable benefits including ease of administration thus improved compliance and lesser cost. The transition to a SQ or oral route of administration would have a considerable effect on treatment feasibility and adherence, thus greatly reducing the chance of breakthrough bleeds related to missed infusions.

Also, the half-life of the current FVIII EHL products ranges from 19 to 22 hours, thus necessitating two weekly IV injections. Thus, despite the advantages of EHL-FVIII, the improvements of pharmacokinetics were only modest at the best. The reason of this is described by the fact that the EHL-FVIII molecules bind with endogenous VWF to form FVIII-VWF complex, that is cleared and has a relatively short half-life (~15 hours) [Citation93]. To counteract this problem, in development is a novel FVIII with VWF fusion, Efanesoctocog Alfa (BIVV001), has an extended half-life (higher dose) of up to 42.5 hours. After seven days of injection, the mean factor activity at one week after infusion was 17% [Citation94].

Now 5 years after FDA approval in the US, emicizumab is considered a “standard of care” product for prophylaxis in patients with HA [Citation14]. A similar agent in development is Mim 8, another bispecific antibody like emicizumab, but in comparison with emicizumab, Mim8 normalized thrombin generation and clot formation, with potencies 13 and 18 times higher than a sequence-identical analog of emicizumab [Citation95].

Additional prospective agents that pursue to target natural anticoagulants thus “rebalancing” a hemorrhagic tendency by introducing a thrombotic tendency are in various stages of development.

Tissue factor pathway inhibitor (TFPI) is a natural anticoagulant that prevents unrestricted amplification of the clotting cascade. It acts not only by inhibiting factor Xa but also inhibits factor VIIa, thereby preventing the generation of factor Xa by the extrinsic pathway [Citation16]. Multiple monoclonal antibodies targeting TFPI are in various stages of development [Citation16,Citation17].

Antithrombin is a natural anticoagulant, a serine protease inhibitor (SERPIN) that inhibits factor IIa, factor Xa, and other serine proteases such as factor IXa, FVIIa, and FXI. Reduced AT activity can lead to a prothrombotic state. Fitusiran is an investigational agent targeting AT [Citation16,Citation18].

Factor V Leiden mutation is a point mutation in the FV gene, which encodes the factor V protein and confers resistance to APC mediated FVa degradation. Over two decades ago, it was observed that the presence of this mutation in hemophiliacs is beneficial to control bleeding, thus hypothesizing the presence of Factor V Leiden mutation as a target in the management of hemophilia [Citation96].

The next incremental improvement in factor replacement therapy may be in the form of factor VIII and IX gene delivery. Gene therapy for hemophilia offers the potential to restore normal or near normal hemostatic function while relieving the problem of frequent infusions with FVIII or FIX concentrates [Citation97]. In hemophilia, gene therapy includes introduction of either a wild-type factor gene or a modified gene with enriched properties such as greater activity or longer half-life. Hepatocytes are an appropriate target cell for transduction since liver is the site of synthesis of most clotting factors. Diverse technologies have been used to introduce the F8 or F9 gene, either using ex vivo genetic modification followed by implantation of the modified cells or by means of a vector carrying a construct that introduces the normal gene or vectors that produce other coagulation factors such as factor VIIa that can be used in hemophiliacs with immune tolerance [Citation21,Citation22]. Adeno-associated viruses (AAV) as vectors are the most frequently used since they do not cause infectious disease in humans, generally do not integrate into the native nuclear genome and are replication-deficient thus decreasing the risk of insertional mutagenesis, Promising findings have been reported from multiple clinical trials [Citation98–100]. The non-integrating nature of AAV vectors leads to low risk of genotoxicity and long-term carcinogenesis but implies some loss of efficacy with replication of the transduced hepatocytes [Citation101]. Despite improved outcomes, several challenges remain, including the presence of pre-existing neutralizing antibodies to the AAV capsids and the unpredictability of transgene expression [Citation102].

So far, data on toxicity leaves unanswered questions, as studies have demonstrated hepatotoxicity with increased transaminases that may affect transgene expression. There is a theoretical risk of late genotoxicity as some vectors (like retroviruses), due to insertional mutagenesis. that may affect naturally occurring tumor suppressor genes. Even though adeno-associated virus vectors have a lower risk of genomic insertion, the ultra high number of vectors required may escalate that risk. To date, one case each of tonsillar carcinoma, acinic cell carcinoma, and hepatocellular carcinoma have been reported in patients with hemophilia post-gene therapy. In all cases, there was little to no evidence of AAV genome integration [Citation103,Citation104] Cellular therapy, another contemporary technique to manage hemophilia is under development that comprises introducing intact cells rather than manipulation of coagulation factor genes. Autologous cells may be treated with a gene construct ex-vivo and then re-introduced [Citation23] .

3.1 Half-life extension

Efanesoctocog alfa (BIVV001, rFVIIIFc-VWF-XTEN, Sanofi, and Sobi) is a novel recombinant FVIII molecule that uses innovative techniques of Fc fusion to the D’D3-domain of vWF and the two XTEN polypeptides to extend its time in circulation. This overrides the dilemma of native VWF mediated clearance, and provides and additional approach to half-life extension [Citation105,Citation106]. In June 2022, the U.S. FDA granted Fast Track Designation for BIVV001in patients with HA [Citation107]. The mean half-life was three to four times as long as that of recombinant FVIII. The dose is 50–65 IU/kg once weekly, and the half-life varies from 37 to 41 hours. In the initial phase 1–2a open-label trial, 16 patients (18 to 65 years of age) with severe HA were assigned to receive BIVV001. No inhibitors to FVIII were detected and no hypersensitivity or anaphylaxis events were reported [Citation108]. A single IV injection of BIVV001 resulted in high sustained FVIII activity levels. Efanesoctocog alfa is currently being evaluated in the ongoing Phase 3 XTEND-1, an open-label, non-randomized interventional study in PTPs ≥12 years of age [n = 150] with severe HA [Citation107].

3.2 Subcutaneous and oral replacement agents

Even though in late 1990s, the administration of SQ FVIII was reported, many animal studies suggested only limited bioavailability of 5 to 10% [Citation109]. It was later revealed that the cause of poor bioavailability was its inactivation in the SQ tissue after binding to phospholipids leading to its degradation [Citation110]. In later studies, it was determined that VWF protects FVIII from proteolytic degradation by C3 domain binding to the C1 domain of FVIII and the best bioavailability was obtained with vWF-12 [Citation111,Citation112]. Various studies have confirmed superior bioavailability up to 41% when rFVIII was co-administered with vWF-12 as opposed to 2% with rFVIII alone [Citation113].

Based on the encouraging preclinical trials, a phase 1, double-blinded, multicenter trial of Turoctocog alfa pegol (SQ N8-GP, Novo Nordisk) in 26 PTPs with severe HA was conducted. However, it was associated with a high incidence of neutralizing antibodies in pretreated patients with severe HA. Hence, further clinical development of SQ N8-GP was suspended [Citation114]. Another agent is SubQ-8 that uses a novel approach for SQ delivery of the recombinant FVIII (simoctocog alfa, Nuwiq) co-administered with a recombinant human VWF fragment dimer that contains the D’D3 domain of VWF that augments bioavailability. The safety and efficacy of SubQ-8 in patients with severe HA is under investigation in a phase 1 and 2 study (NCT04046848) [Citation115].

rFIXFc-XTEN (BIVV002, Sanofi) is a rFIXFc fusion protein that was studied in HB mice that uses recombinant XTEN conjugates to enhance efficacy via SQ injection compared with IV dosing [Citation116]. Another FIX variant is Dalcinonacog alfa (formerly CB 2679d; also designated as ISU 304, Catalyst) which is an rFIX with better properties, including resistance to antithrombin inhibition and increased affinity for FVIIIa. It has a 22-fold potency and 8-fold prolongation of plasma activity than native human FIX as seen in animal and in vitro studies [Citation117]. This was evaluated in a phase 1/2 trial in which there are multiple cohorts, wherein the highest tier of the multidose SQ dosing cohort, the patients received a single loading dose of IV 75 IU/kg, followed by nine daily SQ doses of 150 IU/kg. There were two patients in this cohort. The two patients achieved through FIX levels of 34% and 31%. Unfortunately, 1 subject showed the presence of a non-neutralizing antibody and a neutralizing antibody was identified in a second subject [Citation118]. A phase 2 study demonstrated that SQ dalcinonacog alfa is effective in raising FIX levels comparable to IV EHL rFIX [Citation119].

An overarching and common theme that seems to plague the successful development of SQ factor VIII or IX products is the development of neutralizing antibodies. This suggests that the processing or alteration of the agent in the pathway from the SQ to intravascular space is creating immunogenic epitopes on the product that would not occur if delivered intravenously, directly.

Another agent that was being developed was activated marzeptacog alfa (MarzAA, Catalyst) administered SQ which is a variant of rFVIIa used for the management of hemophilia with inhibitors. It is a recombinant activated human factor VII with four site-specific amino acid changes, two within the catalytic domain to enhance procoagulant activity and two to increase the terminal half-life [Citation120]..

Production of SQ FVIII and FIX, MarzAA, Dalcinonacog alfa (Catalyst) and BIVV002 (Sanofi) and has been halted, however other pharmaceutical companies have SQ agents, such as simoctocog alfa in phase 1/2 trials [Citation121].

Finally, the oral administration of replacement agents is in development. But, achieving functional preservation and bioavailability remains a challenge. The first preliminary, preclinical safety and efficacy data of orally administered rFVIII compared it to a similar dose of FVIII 150 IU/kg administered IV or intraperitoneally. This early study suggested that the orally delivered FVIII restored hemostasis in HA dogs [Citation122]. Because of the technical challenges, it was redirected and found to be effective for oral tolerization. The cholera toxin B subunit fused with FIX is stable in commercially prepared lyophilized lettuce cells [Citation123]. When these lettuce cells were fed to mice with HB, they stimulated latency-associated peptide 1 regulatory T cells that suppressed inhibitor/IgE formation and anaphylaxis against FIX. This was further confirmed in another study including dogs with HB wherein the CTB FIX–expressing lettuce cells led to the suppression of IgG/inhibitor and IgE formation against the IV-administered FIX in 3 of 4 dogs, which led to the marked shortening of coagulation times by IV FIX in the orally tolerized dogs [Citation124].

3.3 Bispecific antibody

Mim8 is a fully human bispecific antibody under development that substitutes the role of FVIII by bridging FIXa and FX on the phospholipid surface of activated platelets, enhancing the proteolytic activity of FIXa, and thus facilitating effective FX activation. It can also be administered SQ in individuals with HA, both with and without inhibitors [Citation95]. In a nonclinical safety and pharmacodynamics study in cynomolgus monkeys, Mim8 was well tolerated in doses ranging from 0.3 to 60 mg/kg/week IV or SQ and thrombosis-related findings were observed in doses above 6 mg/kg/week. Based on the encouraging in vitro and animal studies [Citation125]. Phase 1 and Phase 2 clinical trials are underway (NCT04204408).

3.4 Rebalancing agents: anti-TFPI

Befovacimab (BAY 1093884, Bayer) is a monoclonal antibody against TFPI, an IgG2 fully human monoclonal antibody that binds to both the Kunitz 1 (K1) and Kunitz 2 (K2) domains of the TFPI was not further pursued after three individuals developed central nervous system thromboses [Citation16]. Concizumab (mAb2021, NovoNordisk) is a humanized IgG4 monoclonal antibody against TFPI that targets the K2 domain of the TFPI, that can be administered either via SQ or IV route. It is administered SQ once daily at a starting dose of 0.15 mg/kg and increased as needed or a single IV dose of 0.5–9,000 µg/kg [Citation16]. The safety and efficacy were established in a randomized trial involving 53 patients that included 36 with HA, 9 with HA and inhibitors, and 8 with HB with inhibitors. The patients treated with concizumab prophylaxis versus on-demand rFVIIa demonstrated an 80–90% greater reduction in bleeding than seen with on-demand rFVIIa (ABR with concizumab, 4.5, versus 20.4 with rFVIIa) [Citation17]. Another ongoing phase 3 program evaluating concizumab was put on hold due to thrombotic events, and then the study was restored after a risk mitigation strategy was established [Citation16].. Across both the concizumab phase 2 trials, 7/25 in explorer 4 and 13/36 in explorer 5 receiving daily concizumab prophylaxis underwent 1 or more surgeries during the 18 and 22 months, respectively, and excluded patients requiring major surgery. The number of surgery-related bleeds was low and classified as mild or moderate, with one exception [Citation126].

Marstacimab [PF-06741086, Pfizer] is another human monoclonal antibody IgG1 that binds to the K2 domain of TFPI that can also be administered SQ or IV [Citation16]. In an ongoing trial 26 individuals with HA or HB were treated with marstacimab for six months. The mean ABRs were reduced by 84.5% to 92.6% [Citation127] No complications were reported. MG1113 (Greencross) is humanized IgG4 monoclonal antibody that binds to the Kunitz-2 domain [K2] of TFPI that in preclinical studies has been shown to restore thrombin generation in FVIII or FIX deficiency with and without inhibitors [Citation128]. Both IV and SQ injection resulted in a decreased blood loss from cuticle injury. A pharmacological profile study showed a non-linear pharmacokinetic profile at the dosing range from 2.5 to 10 mg/kg [Citation129].

3.5 Rebalancing agents: reducing AT

Fitusiran (ALN‐AT3, Sanofi) is a small-molecule RNA interference agent that acts by binding to and degrading the mRNA (SERPINC1 gene) encoding AT. It can be administered SQ. In a phase 1 dose escalation study of 25 individuals with moderate to severe HA or HB treated with fitusiran, the number of bleeding events decreased [mean ABR, 0 versus 3] and the effect appeared to be dose-dependent [Citation15]. The drug has short half-life in the plasma [three to five hours]; however, the AT level reduction persists for several weeks [Citation15]. Conversely, in an extension trial, a patient developed a fatal cerebral sinus thrombosis and the study was temporarily halted. It was then restarted with decreased dose intensity following implementation of a risk mitigation strategy [Citation130,Citation131]. A follow-up study did not report thrombotic complications [Citation132]. Nonetheless, it is important to note that if there is a prothrombotic stimulus such as severe injury or infection (such as coronavirus disease 2019 (COVID-19)), that could further potentiate prothrombotic risk.

The initial results from ATLAS A/B study in patients with severe HA and HB with or without inhibitor receiving fitusiran 80 mg once a month compared to daily factor prophylaxis, showed a 90% reduction in ABR in patients receiving fitusiran and no thrombotic events were reported [Citation133]. Dose escalation studies to target AT levels to less than 35% by starting with a dose of 50 mg once in 2 months, if needed escalation to once a month and further to 80 mg once a month [Citation134].

3.6 Modified inhibitors of APC

Apha-1-antitrypsin, plasminogen activator inhibitor 1 and protein C inhibitor are naturally occurring APC inhibitors that are members of the serpin (serine protease inhibitor) family [Citation135,Citation136]. SerpinPC (ApcinteXLtd) which a modification of these serpins was revealed to promote thrombin generation in vitro and improved hemostasis in a murine model of HA [Citation137]. Presently, a phase 1/2 trial (NCT04073498) of Serpin PC is in progress in hemophiliacs and healthy volunteers. Additionally, a monoclonal antibody to APC (HAPC1573) that exhibited encouraging results in a primate study [Citation138].

Besides the reassuring results, it is important to note that AT and APC also has roles in inflammatory signaling and consequently there are concerns that interference of APC function could have effects beyond the coagulation cascade. To evade the other undesirable effects of APC inhibition, modified versions of natural serpins are under investigation that specifically block APC activity in the coagulation cascade without disrupting other pathways.

Protein S acts as a cofactor for APC. A monoclonal antibodies that inhibits protein S and and siRNA knockdown demonstrating increased thrombin generation and resistance to APC and TFPI in murine models are in early stages of development [Citation139].

3.7 Gene Therapy

Valoctocogene roxaparvovec (valrox) for HA showed clinically meaningful improvements in bleeding rates with sustained increase in FVIII activity [Citation99]. No development of inhibitors or thrombosis noted [Citation99]. Patients with anti-AAV5 capsid antibodies and substantial liver dysfunction were excluded from this trial [Citation99]. Valrox (Roctavian in Europe) is a codon-optimized adeno-associated virus serotype 5 (AAV5) vector containing a gene for B-domain deleted human FVIII (AAV5-hFVIII-SQ) which in turn is a non-integrating hepatotropic vector that remains mostly episomal [Citation140]. The 6-year follow-up data of the phase 1/2 trial 5 showed a sustained reduction in the ABR. Twelve out of 13 patients did not require FVIII prophylaxis [Citation141]. The phase 3 trial GENEr8-1 demonstrated an 85% decline in ABR and 98% decline in the annual FVIII utilization [Citation99]. The proportion of zero bleeds (52.7% versus 28.5%; p < 0.05) or zero treated bleeds (79.5% vs 32.9%; p < 0.001) was significantly high with the gene therapy in a post hoc analysis. No liver damage was seen in the five patients of a phase I/II trial that consisted of performing a liver biopsy upto 4 years after treatment [Citation142]. There was no clonal expansion seen and even though one to six integration events per 1000 cells were observed, there was no enrichment in integration sites near the proto-oncogene. Another adenovirus-based gene therapy that is currently being investigated in a phase I/II study in patients with HA is TAK754 which is AAV8 based [Citation143]. On August 2022, the European commission granted Valoctocogene roxaparvovec (BioMarin Pharmaceutical Inc.), conditional marketing authorization for the treatment of severe HA in adult patients without a history of FVIII inhibitors and without detectable antibodies to AAV5 thus launching the commercial availability of gene therapy [Citation144].

For HB F9 Padua variant containing therapy that uses a AAV5 vector is Etranacogene dezaparvovec (Hemgenix). On November 22nd, 2022, the U.S. FDA approved Hemgenix, for the treatment of adults with HB who currently use FIX prophylaxis therapy [Citation145].

This therapy has been studied in HOPE-B, a phase III trial in adults with severe or moderately severe HB and has demonstrated a significant improvement in quality of life and reduction in treatment burden (nominal p value <0.0001) [Citation146]. The safety and efficacy data in 54 participants over a period of 24 months with or without pre-existing AAV5 neutralizing antibodies showed the mean ABR was significantly reduced by 64% (mean ABR 1.51; p = 0.0002), there was 96% reduction in annualized FIX consumption and the treatment was well tolerated with no serious AEs [Citation147].

Another gene therapy for HB, the F9 Padua variant (F9 p.R338L) contains a naturally occurring missense mutation in the F9 gene that increases its activity approximately 4 to 40-fold and has been most widely used for FIX gene therapy trials [Citation148,Citation149]. A recent phase 1–2 data B-LIEVE established the safety and efficacy of Verbrinacogene setparvovec (FLT180a) in patients with severe or moderately severe HB which is a liver-directed AAV gene therapy that uses a synthetic capsid and a gain-of-function protein to normalize FIX levels. This trial reported that the first three patients achieved normal FIX levels and discontinued prophylaxis by days 33, 56 and 77 and no serious adverse effects were observed [Citation150].

Considering the potential theoretical risk of genotoxicity of viral vector mediated gene transfer, other non-viral means of gene therapy are being explored. One such technique was ultrasound mediated gene delivery, and animal study showed promising results. In this study, the portal vein of mice was injected with plasmid DNA and pulsed therapeutic ultrasound transducer was applied to the liver [Citation151]. Another successful animal study, in mice with HA, demonstrated delivery of lentiviral vectors into bone marrow to drive FVIII expression in platelets [Citation152]. Lipid nanoparticle technology to correct frameshift deletion mutation in exon 1 of FVIII gene by delivering a targeted single guide RNA with CRISPR/Cas9-based gene editing was recently shown to achieve therapeutic levels of FVIII expression [Citation153]. In patients with HB with inhibitors who fail immune tolerance induction, CRISPR/Cas9 expressing plasmid may be a useful method to insert a transgene encoding murine FVIIa into the host genome [Citation154]. Inhibition of antithrombin pathway via nanobodies packaged in an AAV8 vector to control bleeding in patients with HA has also been studied in mice [Citation155].

3.8 Cellular Therapy

Multiple preclinical studies have supported the transduction of hematopoietic stem cells ex vivo with lentiviruses to generate genetically modified platelets to control bleeding in patients with severe HA with inhibitors [Citation156,Citation157].

Hepatocyte transplantation has been studied in mouse model of HA. The study implied that the transplantation of liver sinusoidal endothelial cells from non-hemophilic donor animals resulted in increased factor levels and correction of the bleeding phenotype (23). SIG-001 is a polymer-encapsulated human cell system to express FVIII that was not pursued due to the development of a FVIII inhibitor in one out of three patients [Citation158].

4. Conclusion

In this expert review, we explore the past, present, and future directions in the treatment of people living with Hemophilia A and B. The new innovative therapies could be revolutionary based on the cost and availability of these products. Though optimistic, one cannot apprehend what the future holds in the next decade or two. Whether it was thousands of people contracting HIV before concentrates were rendered safe or thrombotic risk with the newer agents and more so recently a rather uncharted territory of gene modification, caution must be implemented. Despite all this, the benefits that were not expected with prior therapies are a reality for many. However, more than three-fourths of the world’s hemophilia population still do not get the standard therapy. With the sustained efforts by the World Federation of Hemophilia to provide harmonization of health services worldwide, we still see light at the end of the tunnel and hope to achieve consistency in hemophilia management. With continued innovation in the treatment approaches, the clinician and patient need to make an informed decision individualizing priority and prerequisites. With current ongoing clinical trials, we expect that clinicians with their patients can tailor management assessing phenotype, patient goals both for education and vocation and thus provide optimal therapy so that we can make the life of hemophiliac patients more integrated into the framework of society.

5. Expert opinion

Current therapies in the management of hemophilia have demonstrated safety and efficacy over decades of data but the future holds better promise. Numerous techniques have been developed to optimize safety and effectiveness of the existing plasma products. The availability of safer plasma-derived products and recombinant factor concentrates helped improve joint outcomes and quality of life. Nevertheless, the real-world downsides of clotting factor infusions include reliance on venous access, poor adherence, and scarce affordability on a global scale. The advent of EHL product therapies has further enhanced prophylaxis effectiveness and compliance and is being used worldwide through the implementation of humanitarian programs by the World Federation of Hemophilia aiming at providing such therapies to socio-economically disadvantaged countries albeit the task remains challenging. Despite the major advances of these EHL products, breakthrough bleeding, IV administration, inhibitor development and the need for treatment individualization still pose major challenges to the clinician and the patients. Also, though the half-lives of rIX products have been extended by 3–6 folds but the half-life prolongation of rVIII products is only roughly 1.5–1.6 folds. In light of these challenges, the substitution therapy/non-factor therapies (emicizumab) and hemostatic rebalancing therapies bring several few advantages. Since they do not expose the patient to FVIII or FIX to elicit inhibitors, the non-replacement therapies function despite existing inhibitors. The SQ route of administration and the availability of fixed-dose regimens are additional benefits. These therapies also offer the potential to avoid exposure to the immunogenic proteins or to explore more tolerogenic approaches to introduction of the factor replacement therapies in cases of breakthrough bleeding. Thus, non-factor agents may also be useful for prophylaxis from an early age and even from birth.

To circumvent existing concerns in the management of hemophilia, novel therapies are underway. Recombinant FVIII products with extended half-life might prove beneficial without substantially affecting the therapeutic intervals. The use of innovative non-replacement products could be an alternative strategy in patients both with and without inhibitors though long-term data and safety remains a concern. SQ FVIII and FIX agents with the desirability of ease of administration are currently being explored under various stages of clinical trial. Also, animal studies for oral administration of factors have been encouraging. Gene therapy has so far demonstrated a robust and resilient expression of efficacious levels of FVIII and FIX with about half a decade worth of data. Recognized and unfamiliar risks associated to immunologic responses, cellular stress induction, and genome integration remain. Few studies have demonstrated cancer risk; however, a proven association and further long-term data remain to be unraveled. Also, inconsistency of gene expression has generated some ambiguity. Ongoing careful monitoring, investigation long-term data and its application in pediatric population remains to be explored.

Article highlights

  • Factor replacement products once the mainstay of hemophilia treatment, however newer products are now available with easier modes of administration, decreased immunogenicity and increased efficacy.

  • Recombinant bioengineering with the development of extended half-life replacement therapies are available to diminish the frequency of administration and improved compliance.

  • Emicizumab is considered by many to be another standard of care agent for bleed prophylaxis in patients with hemophilia A.

  • Subcutaneous administration of some these new therapies makes it further attractive for prophylaxis in patients with poor venous access and studies are underway to develop subcutaneous and even oral replacement therapies.

  • The most striking of newer development is gene therapy that involves the introduction of either a wild-type or a modified factor gene with greater activity or longer half-life and establishing continuous endogenous expression of factor VIII or factor IX.

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Abbreviations

HA Hemophilia A HB Hemophilia B FVIII Factor VIII and FIX Factor IX TFPI Tissue Factor Pathway Inhibitor SHL Standard half-life EHL Extended half life; IV intravenous; SQ subcutaneous PEG polyethylene glycol FDA Food and Drug Administration ABR annualized bleeding rates vWF Von Willebrand factor AAV adeno-associated virus dL deciliter IU international units aPCC activated prothrombin concentrate PUPs previously untreated patients PTPs previously treated patients

Additional information

Funding

The paper was not funded.

References

  • Mannucci PM, Tuddenham EGD. The Hemophilias — From Royal Genes to Gene Therapy. N Engl J Med. 2001 Jun 7;344(23):1773–1779.
  • Accessed on 2022 Sep 4. https://www.hemophilia.org/bleeding-disorders-a-z/overview/history
  • Schramm W. The history of haemophilia - a short review. Thromb Res. 2014 Nov;134(Suppl 1):S4–9.
  • Mannucci PM. Hemophilia and related bleeding disorders: a story of dismay and success. In: Hematology Am Soc Hematol Educ Program. ASH. 2002. p. 1–9.
  • Wight J, Paisley S. The epidemiology of inhibitors in haemophilia A: a systematic review. Haemophilia. 2003 Jul;9(4):418–435.
  • Volkers P, Hanschmann KM, Calvez T, et al. Recombinant factor VIII products and inhibitor development in previously untreated patients with severe haemophilia A: Combined analysis of three studies. Haemophilia. 2019 May;25(3):398–407.
  • Male C, NG A, Rafowicz A, et al. Inhibitor incidence in an unselected cohort of previously untreated patients with severe haemophilia B: a PedNet study. Haematologica. 2021 Jan 1;106(1): 123–9.
  • Valentino LA, Kempton CL, Kruse-Jarres R, et al. US Guidelines for immune tolerance induction in patients with haemophilia a and inhibitors. Haemophilia. 2015 Sep;21(5):559–567.
  • Di Minno G, Castaman G, de Cristofaro R, et al. Progress, and prospects in the therapeutic armamentarium of persons with congenital hemophilia. Defining the place for liver-directed gene therapy. Blood Rev. 2022 Aug;23:101011.
  • Sherman A, Biswas M, Herzog RW. Innovative Approaches for Immune Tolerance to Factor VIII in the Treatment of Hemophilia A. Front Immunol. 2017;8:1604.
  • Reipert BM, Gangadharan B, Hofbauer CJ, et al. The prospective Hemophilia Inhibitor PUP Study reveals distinct antibody signatures prior to FVIII inhibitor development. Blood Adv. 2020 Nov 24;4(22):5785–5796.
  • AD S, IS M, Nasr S, et al. The future of bypassing agents for hemophilia with inhibitors in the era of novel agents. J Thromb Haemost. 2018 Dec;16(12): 2362–74.
  • Matino D, Makris M, Dwan K, et al. Recombinant factor VIIa concentrate versus plasma-derived concentrates for treating acute bleeding episodes in people with haemophilia and inhibitors. Cochrane Database Syst Rev. 2015 Dec 16;2015(12):CD004449.
  • Langer AL, Etra A, Aledort L. Evaluating the safety of emicizumab in patients with hemophilia A. Expert Opin Drug Saf. 2018 Dec;17(12):1233–1237.
  • Pasi KJ, Rangarajan S, Georgiev P, et al. Targeting of Antithrombin in Hemophilia A or B with RNAi Therapy. N Engl J Med. 2017 Aug 31;377(9):819–828.
  • Mahlangu JN. Progress in the Development of Anti-tissue Factor Pathway Inhibitors for Haemophilia Management. Front Med (Lausanne). 2021;8:670526.
  • Shapiro AD, Angchaisuksiri P, Astermark J, et al. Subcutaneous concizumab prophylaxis in hemophilia A and hemophilia A/B with inhibitors: phase 2 trial results. Blood. 2019 Nov 28;134(22):1973–1982.
  • https://hemophilianewstoday.com/2021/03/01/fitusiran-dosing-phase-3-trials-reduced-to-lower-risk-of-blood-clots (Accessed on 2022 Sep 4th).
  • Aledort L, Mannucci PM, Schramm W, et al. Factor VIII replacement is still the standard of care in haemophilia A. Blood Transfus. 2019 Nov;17(6):479–486.
  • Santagostino E, Martinowitz U, Lissitchkov T, et al. Long-acting recombinant coagulation factor IX albumin fusion protein (rIX-FP) in hemophilia B: results of a phase 3 trial. Blood. 2016 Apr 7;127(14):1761–1769.
  • Nathwani AC. Gene therapy for hemophilia. Hematology Am Soc Hematol Educ Program. 2019 Dec 6;2019(1):1–8.
  • Margaritis P, Roy E, Aljamali MN, et al. Successful treatment of canine hemophilia by continuous expression of canine FVIIa. Blood. 2009 Apr 16;113(16):3682–3689.
  • Trevisan B, Rodriguez M, Medder H, et al. Autologous bone marrow-derived MSCs engineered to express oFVIII-FLAG engraft in adult sheep and produce an effective increase in plasma FVIII levels. Front Immunol. 2022;13:1070476.
  • Nilsson IM, Berntorp E, Löfqvist T, et al. Twenty-five years’ experience of prophylactic treatment in severe haemophilia A and B. J Intern Med. 1992 Jul;232(1):25–32.
  • Berntorp E, Astermark J, Björkman S, et al. Consensus perspectives on prophylactic therapy for haemophilia: summary statement. Haemophilia. 2003 May;9(Suppl 1):1–4.
  • 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 Oct;236(4):391–399.
  • Mannucci PM. AIDS, hepatitis and hemophilia in the 1980s: memoirs from an insider. J Thromb Haemost. 2003 Oct;1(10):2065–2069.
  • Lambert T, Benson G, Dolan G, et al. Practical aspects of extended half-life products for the treatment of haemophilia. Ther Adv Hematol. 2018 9;Sep(9):295–308.
  • Dumont JA, Liu T, Low SC, et al. Prolonged activity of a recombinant factor VIII-Fc fusion protein in hemophilia A mice and dogs. Blood. 2012 Mar 29;119(13):3024–3030.
  • Ivens IA, Baumann A, McDonald TA, et al. PEGylated therapeutic proteins for haemophilia treatment: a review for haemophilia caregivers. Haemophilia. 2013 Jan;19(1):11–20.
  • Gringeri A, Mantovani LG, Scalone L, et al. Cost of care and quality of life for patients with hemophilia complicated by inhibitors: the COCIS Study Group. Blood. 2003 Oct 1;102(7):2358–2363.
  • Mannucci PM. Hemophilia therapy: the future has begun. Haematologica. 2020 Mar;105(3):545–553.
  • https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/761083s002s004lbl.pdf (Accessed on 2022 Oct 4).
  • Shapiro AD. Concizumab: a novel anti-TFPI therapeutic for hemophilia. Blood Adv. 2021 Jan 12;5(1):279.
  • Mannucci PM. The choice of plasma-derived clotting factor concentrates. Baillieres Clin Haematol. 1996 Jun;9(2):273–290.
  • US Food and Drug Administration [internet]. https://www.fda.gov/vaccines-blood-biologics/approved-blood-products (cited 2022 Sep 15).
  • Lusher JM. Recombinant clotting factor concentrates. Baillieres Clin Haematol. 1996 Jun;9(2):291–303.
  • Fijnvandraat K, Berntorp E, ten Cate JW, et al. Recombinant, B-domain deleted factor VIII (r-VIII SQ): pharmacokinetics and initial safety aspects in hemophilia A patients. Thromb Haemost. 1997 Feb;77(2):298–302.
  • Schmidbauer S, Witzel R, Robbel L, et al. Physicochemical characterisation of rVIII-SingleChain, a novel recombinant single-chain factor VIII. Thromb Res. 2015 Aug;136(2):388–395.
  • Berntorp E. Second generation, B-domain deleted recombinant factor VIII. Thromb Haemost. 1997 Jul;78(1):256–260.
  • Bray GL, Gomperts ED, Courter S, et al. A multicenter study of recombinant factor VIII (recombinate): safety, efficacy, and inhibitor risk in previously untreated patients with hemophilia A. The Recombinate Study Group. Blood. 1994 May 1;83(9):2428–2435.
  • Seremetis S, Lusher JM, Abildgaard CF, et al. Human recombinant DNA-derived antihaemophilic factor (factor VIII) in the treatment of haemophilia A: conclusions of a 5-year study of home therapy. The KOGENATE Study Group. Haemophilia. 1999 Jan;5(1):9–16.
  • Saxena K, Lalezari S, Oldenburg J, et al. Efficacy and safety of BAY 81-8973, a full-length recombinant factor VIII: results from the LEOPOLD I trial. Haemophilia. 2016 Sep;22(5):706–712.
  • Stasyshyn O, Djambas Khayat C, Iosava G, et al. Safety, efficacy and pharmacokinetics of rVIII-SingleChain in children with severe hemophilia A: results of a multicenter clinical trial. J Thromb Haemost. 2017 Apr;15(4):636–644.
  • White G, Shapiro A, Ragni M, et al. Clinical evaluation of recombinant factor IX. Semin Hematol. 1998 Apr;35(2 Suppl 2):33–38.
  • Roth DA, Kessler CM, Pasi KJ, et al. Human recombinant factor IX: safety and efficacy studies in hemophilia B patients previously treated with plasma-derived factor IX concentrates. Blood. 2001 Dec 15;98(13):3600–3606.
  • Shapiro AD, Di Paola J, Cohen A, et al. The safety and efficacy of recombinant human blood coagulation factor IX in previously untreated patients with severe or moderately severe hemophilia B. Blood. 2005 Jan 15;105(2):518–525.
  • Windyga J, Lissitchkov T, Stasyshyn O, et al. Pharmacokinetics, efficacy and safety of BAX326, a novel recombinant factor IX: a prospective, controlled, multicentre phase I/III study in previously treated patients with severe (FIX level <1%) or moderately severe (FIX level ≤2%) haemophilia B. Haemophilia. 2014 Jan;20(1):15–24.
  • Schiavoni M, Napolitano M, Giuffrida G, et al. Status of Recombinant Factor VIII Concentrate Treatment for Hemophilia a in Italy: Characteristics and Clinical Benefits. Front Med (Lausanne). 2019;6:261.
  • Abshire TC, Brackmann HH, Scharrer I, et al. Sucrose formulated recombinant human antihemophilic factor VIII is safe and efficacious for treatment of hemophilia A in home therapy–International Kogenate-FS Study Group. Thromb Haemost. 2000 Jun;83(6):811–816.
  • Gouw SC, van der Bom JG, Ljung R, et al. Factor VIII products and inhibitor development in severe hemophilia A. N Engl J Med. 2013 Jan 17;368(3):231–239.
  • 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 May 26;374(21):2054–2064.
  • US Food and Drug Administration [internet] ELOCTATE prescribing information. 2017. [Accessed on 2022 Feb 09]. Available at: https://www.fda.gov/downloads/BiologicsBloodVaccines/BloodBloodProducts/ApprovedProducts/LicensedProductsBLAs/FractionatedPlasmaProducts/UCM400192.pdf.
  • 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 Mar 29;119(13):3031–3037.
  • Mahlangu J, Powell JS, Ragni MV, et al. Phase 3 study of recombinant factor VIII Fc fusion protein in severe hemophilia A. Blood. 2014 Jan 16;123(3):317–325.
  • 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 Jun;13(6):967–977.
  • 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 Jan;22(1):72–80.
  • 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 Jul 4;116(1):1–8.
  • 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 Mar;24(2):245–252.
  • Turecek PL, Bossard MJ, Graninger M, et al. BAX 855, a PEGylated rFVIII product with prolonged half-life. Development, functional and structural characterisation. Hamostaseologie. 2012;32(Suppl 1):S29–38.
  • US Food and Drug Administration [internet]. ADYNOVATE prescribing information; 2016. Accessed on 2022 Feb 07. Available at: https://www.fda.gov/downloads/BiologicsBloodVaccines/BloodBloodProducts/ApprovedProducts/LicensedProductsBLAs/FractionatedPlasmaProducts/UCM472594.pdf
  • Gruppo R, López-Fernández 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 Sep;25(5):773–781.
  • 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 Aug 27;126(9):1078–1085.
  • TE C, MT R, JC L, 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 Apr;12(4):488–496.
  • US Food and Drug Administration [internet]. JIVI Antihemophilic Factor (Recombinant) PEGylated-aucl prescribing information; 2018. Accessed on 2022 Jan 11. Available at: https://www.fda.gov/downloads/BiologicsBloodVaccines/UCM618979.pdf
  • 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 Sep;24(5):733–740.
  • 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 Nov;183:13–19.
  • 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 Mar;15(3):411–419.
  • 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 Mar 15;130:11–20.
  • US Food and Drug Administration [internet]. February 19, 2019 Approval Letter - ESPEROCT. BL 125671/0. Accessed on 2022 Oct 10. Available at: https://www.fda.gov/media/120350/download
  • 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 Apr;11(4):670–678.
  • 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 Aug 30;117(9):1705–1713.
  • Peters RT, Low SC, Kamphaus GD, et al. Prolonged activity of factor IX as a monomeric Fc fusion protein. Blood. 2010 Mar 11;115(10):2057–2064.
  • Shapiro AD, Ragni MV, LA V, 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 Jan 19;119(3):666–672.
  • Powell JS, Pasi KJ, Ragni MV, et al. Phase 3 Study of Recombinant Factor IX Fc Fusion Protein in Hemophilia B. N Engl J Med. 2013 Dec 12;369(24):2313–2323.
  • Fischer K, Kulkarni R, Nolan B, et al. Recombinant factor IX Fc fusion protein in children with haemophilia B (Kids B-LONG): results from a multicentre, non-randomised phase 3 study. Lancet Haematol. 2017 Feb;4(2):e75–82.
  • Cooley B, Funkhouser W, Monroe D, et al. Prophylactic efficacy of BeneFIX vs Alprolix in hemophilia B mice. Blood. 2016 Jul 14;128(2):286–292.
  • https://www.fda.gov/media/96526/download. cited 2022 Sep 15.
  • Martinowitz U, Lubetsky A. Phase I/II, open-label, multicenter, safety, efficacy and PK study of a recombinant coagulation factor IX albumin fusion protein (rIX-FP) in subjects with hemophilia B. Thromb Res. 2013 Mar;131(Suppl 2):S11–4.
  • Østergaard H, Bjelke JR, Hansen L, et al. Prolonged half-life and preserved enzymatic properties of factor IX selectively PEGylated on native N-glycans in the activation peptide. Blood. 2011 Aug 25;118(8):2333–2341.
  • Negrier C, Knobe K, Tiede A, et al. Enhanced pharmacokinetic properties of a glycoPEGylated recombinant factor IX: a first human dose trial in patients with hemophilia B. Blood. 2011 Sep 8;118(10):2695–2701.
  • Collins PW, Young G, Knobe K, et al. Recombinant long-acting glycoPEGylated factor IX in hemophilia B: a multinational randomized phase 3 trial. Blood. 2014 Dec 18;124(26):3880–3886.
  • 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 Oct;18(10):1570–1574.
  • https://www.fda.gov/downloads/drugs/guidances/ucm459987.pdf (Accessed on Nov 29, 2022).
  • Shima M, Hanabusa H, Taki M, et al. Factor VIII-Mimetic Function of Humanized Bispecific Antibody in Hemophilia A. N Engl J Med. 2016 May 26;374(21):2044–2053.
  • Oldenburg J, Mahlangu JN, Kim B, et al. Emicizumab Prophylaxis in Hemophilia A with Inhibitors. N Engl J Med. 2017 Aug 31;377(9):809–818.
  • Young G, Liesner R, Chang T, et al. A multicenter, open-label phase 3 study of emicizumab prophylaxis in children with hemophilia A with inhibitors. Blood. 2019 Dec 12;134(24):2127–2138.
  • Mahlangu J, Oldenburg J, Paz-Priel I, et al. Emicizumab Prophylaxis in Patients Who Have Hemophilia A without Inhibitors. N Engl J Med. 2018 Aug 30;379(9):811–822.
  • Klamroth R, Wojciechowski P, Aballéa S, et al. Efficacy of rFVIIIFc versus Emicizumab for the Treatment of Patients with Hemophilia A without Inhibitors: Matching-Adjusted Indirect Comparison of A-LONG and HAVEN Trials. J Blood Med. 2021;12:115–122.
  • Aledort LM. Deaths Associated with Emicizumab in Patients with Hemophilia A. N Engl J Med. 2019 Nov 7;381(19):1878–1879.
  • Linari S, Castaman G. Concomitant Use of rFVIIa and Emicizumab in People with Hemophilia A with Inhibitors: Current Perspectives and Emerging Clinical Evidence. Ther Clin Risk Manag. 2020;16:461–469.
  • 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 May;35:43–50.
  • Pipe SW, Montgomery RR, Pratt KP, et al. Life in the shadow of a dominant partner: the FVIII-VWF association and its clinical implications for hemophilia A. Blood. 2016 Oct 20;128(16):2007–2016.
  • Croteau SE, Wang M, Wheeler AP. 2021 clinical trials update: Innovations in hemophilia therapy. Am J Hematol. 2021 Jan;96(1):128–144.
  • Østergaard H, Lund J, Greisen PJ, et al. A factor VIIIa-mimetic bispecific antibody, Mim8, ameliorates bleeding upon severe vascular challenge in hemophilia A mice. Blood. 2021 Oct 7;138(14):1258–1268.
  • Vianello F, Belvini D, Dal Bello F, et al. Mild bleeding diathesis in a boy with combined severe haemophilia B (C(10400)–>T) and heterozygous factor V Leiden. Haemophilia. 2001 Sep;7(5):511–514.
  • Rodríguez-Merchán EC, de Pablo-Moreno JA, Liras A, et al. Gene Therapy in Hemophilia: Recent Advances. In: Int J Mol Sci. 2021;22:14.
  • George LA, Monahan PE, Eyster ME, et al. Multiyear Factor VIII Expression after AAV Gene Transfer for Hemophilia A. N Engl J Med. 2021 Nov 18;385(21):1961–1973.
  • Ozelo MC, Mahlangu J, Pasi KJ, et al. Valoctocogene Roxaparvovec Gene Therapy for Hemophilia A. N Engl J Med. 2022 Mar 17;386(11):1013–1025.
  • Chowdary P, Shapiro S, Makris M, et al. Phase 1-2 Trial of AAVS3 Gene Therapy in Patients with Hemophilia B. N Engl J Med. 2022 Jul 21;387(3):237–247.
  • Monahan PE, Négrier C, Tarantino M, et al. Emerging Immunogenicity and Genotoxicity Considerations of Adeno-Associated Virus Vector Gene Therapy for Hemophilia. J Clin Med. 2021 Jun 2;10(11):2471.
  • Anti-AAV WT. Antibodies in AAV Gene Therapy: Current Challenges and Possible Solutions. Front Immunol. 2021;17:12.
  • Schmidt M, Foster G R, Coppens M, et al. Liver Safety Case Report from the Phase 3 HOPE-B Gene Therapy Trial in Adults with Hemophilia B [abstract]. Res Pract Thromb Haemost. 2021;5(Suppl 2). Accessed 2023 Jan 6 https://abstracts.isth.org/abstract/liver-safety-case-report-from-the-phase-3-hope-b-gene-therapy-trial-in-adults-with-hemophilia-b/.
  • Rogers GL, Herzog RW. Gene therapy for hemophilia. Front Biosci (Landmark Ed). 2015 Jan 1;20(3):556–603.
  • Schellenberger V, Wang CW, Geething NC, et al. A recombinant polypeptide extends the in vivo half-life of peptides and proteins in a tunable manner. Nat Biotechnol. 2009 Dec;27(12):1186–1190.
  • Seth Chhabra E, Liu T, Kulman J, et al. BIVV001, a new class of factor VIII replacement for hemophilia A that is independent of von Willebrand factor in primates and mice. Blood. 2020 Apr 23;135(17):1484–1496.
  • FDA grants priority review to efanesoctocog alfa for people with hemophilia A. pdf (Accessed 2022 Nov 3).
  • Konkle BA, Shapiro AD, Quon DV, et al. BIVV001 Fusion Protein as Factor VIII Replacement Therapy for Hemophilia A. N Engl J Med. 2020 Sep 10;383(11):1018–1027.
  • Spira J, Widlund L, Österberg T, et al. Pharmaceutical formulation for subcutaneous, intramuscular or intradermal administration of factor VIII of factor IX. International patent application: US patent US5925739A, European patent EP0772452. 1995 March 30. https://patentimages.storage.googleapis.com/ec/59/6c/cfe13d59588396/US5925739.pdf
  • Shi Q, Kuether EL, Schroeder JA, et al. Intravascular recovery of VWF and FVIII following intraperitoneal injection and differences from intravenous and subcutaneous injection in mice. Haemophilia. 2012 Jul;18(4):639–646.
  • Sadler JE. New concepts in von Willebrand disease. Annu Rev Med. 2005;56:173–191.
  • Fatouros A, Lidén Y, Recombinant Factor SB. VIII SQ—Stability of VIII: C in Homogenates from Porcine, Monkey and Human Subcutaneous Tissue. J Pharm Pharmacol. 2010 Feb 18;52(7):797–805.
  • Vollack-Hesse N, Oleshko O, Werwitzke S, et al. Recombinant VWF fragments improve bioavailability of subcutaneous factor VIII in hemophilia A mice. Blood. 2021 Feb 25;137(8):1072–1081.
  • Klamroth R, Feistritzer C, Friedrich U, et al. Pharmacokinetics, immunogenicity, safety, and preliminary efficacy of subcutaneous turoctocog alfa pegol in previously treated patients with severe hemophilia A (alleviate 1). J Thromb Haemost. 2020 Feb;18(2):341–351.
  • Solecka-Wituska BA, Wuschko S, Kannicht C. Pharmacokinetics of subcutaneously administered simoctocog alfa with von Willebrand factor fragment candidates in a minipig model [abstract]. In: Disorders of coagulation or fibrinolysis. Blood. 2017;130(9):1073.
  • Liu Z, van der Flier A, Ismail A, et al. Evaluation of Recombinant FIXFc-XTEN Bleeding Efficacy in Hemophilia-B Mouse Models [abstract]. Blood. 2016;128(22):Abstract 3757.
  • Csh WY, Levy H, Lee M, et al. Phase 1/2 trial of subcutaneously administered Factor IX variant CB2679d/ISU304: pharmacokinetic and activity [abstract]. Blood. 2017;130(suppl1):Abstract 87.
  • Catalyst Biosciences provides update on CB 2679d/ISU304 factor IX clinical program in Hemophilia B. http://ir.catalystbiosciences.com/phoenix.zhtml. cited 2022 Oct 19
  • Mahlangu J, Levy H, Lee M, et al. Efficacy and safety of subcutaneous prophylaxis with dalcinonacog alfa in adults with haemophilia B. Haemophilia. 2021 Jul;27(4):574–580.
  • Mahlangu J, Levy H, Kosinova MV, et al. Subcutaneous engineered factor VIIa marzeptacog alfa (activated) in hemophilia with inhibitors: Phase 2 trial of pharmacokinetics, pharmacodynamics, efficacy, and safety. Res Pract Thromb Haemost. 2021 Aug 17;5(6):e12576.
  • Mancuso ME, Mahlangu JN, Pipe SW. The changing treatment landscape in haemophilia: from standard half-life clotting factor concentrates to gene editing. Lancet. 2021 Feb 13;397(10274):630–640.
  • Hashim MA, Yamaguchi A, Vo AT, et al. Orally delivered factor VIII restores hemostasis in a hemophilia A dog. ISTH 2021 Congress. Abstract number: OC 48.4.
  • Su J, Zhu L, Sherman A, et al. Low cost industrial production of coagulation factor IX bioencapsulated in lettuce cells for oral tolerance induction in hemophilia B. Biomaterials. 2015 Nov;70:84–93.
  • Herzog RW, Nichols TC, Su J, et al. Oral Tolerance Induction in Hemophilia B Dogs Fed with Transplastomic Lettuce. Mol Ther. 2017 Feb 1;25(2):512–522.
  • Lauritzen B, Bjelke M, Björkdahl O, et al. A novel next‐generation FVIIIa mimetic, Mim8, has a favorable safety profile and displays potent pharmacodynamic effects: Results from safety studies in cynomolgus monkeys. J Thromb Haemost. 2022 Jun 6;20(6):1312–1324.
  • Wheeler AP, Benson G, Eichler H, et al. Surgeries and diagnostic procedures in hemophilia patients on concizumab prophylaxis: results from the phase 2 Explorer4 and Explorer5 trials [abstract]. In: Disorders of coagulation or fibrinolysis: Clinical and Epidemiological. Blood. 2021;138(Suppl 1):345.
  • Mahlangu J, Lamas JL, Morales JC, et al. Long-term Safety and Efficacy of the Anti-TFPI Monoclonal Antibody Marstacimab in Patients with Severe Haemophilia A or B: Results from a Phase 2 Long-term Treatment Study [abstract]. 2021. https://abstracts.isth.org/abstract/long-term-safety-and-efficacy-of-the-anti-tfpi-monoclonal-antibody-marstacimab-in-patients-with-severe-haemophilia-a-or-b-results-from-a-phase-2-long-term-treatment-study/
  • Kwak H, Lee S, Jo S, et al. MG1113, a specific anti–tissue factor pathway inhibitor antibody, rebalances the coagulation system and promotes hemostasis in hemophilia. Res Pract Thromb Haemost. 2020 Nov 22;4(8):1301–1312.
  • Kwak E, Kim MJ, Park JH, et al. Target‐mediated drug disposition modeling of an anti‐TFPI antibody (MG1113) in cynomolgus monkeys to predict human pharmacokinetics and pharmacodynamics. J Thromb Haemost. 2021 Jun 17;19(6):1425–1435.
  • http://investors.alnylam.com/releasedetail.cfm?ReleaseID=1039464 (Accessed on 2022 Sep 07
  • https://hemophilianewstoday.com/2021/03/01/fitusiran-dosing-phase-3-trials-reduced-to-lower-risk-of-blood-clots (Accessed on 2022 Nov 18
  • Pasi KJ, Lissitchkov T, Mamonov V, et al. Targeting of antithrombin in hemophilia A or B with investigational siRNA therapeutic fitusiran-Results of the phase 1 inhibitor cohort. J Thromb Haemost. 2021 Jun;19(6):1436–1446.
  • Srivastava A, Rangarajan S, Kavakli K, et al. Fitusiran, an investigational siRNA therapeutic targeting antithrombin for the treatment of hemophilia: first results from a phase 3 study to evaluate efficacy and safety in people with hemophilia A or B without inhibitors (ATLAS-A/B). ASH abstract. Blood. 2021;138(Suppl2):LBA–3.
  • Bhagunde P, Ge S, Iqbal S, et al. Fitusiran population pharmacokinetic and pharmacodynamic (PopPK/PD) modeling to support revised dose, dosing regimens & dose mitigation scheme. ISTH abstract. Res Pract Thromb Haemost. 2021;5(Suppl):2.
  • Rezaie AR. Vitronectin functions as a cofactor for rapid inhibition of activated protein C by plasminogen activator inhibitor-1. Implications for the mechanism of profibrinolytic action of activated protein C. J Biol Chem. 2001 May 11;276(19):15567–15570.
  • Suzuki K, Nishioka J, Kusumoto H, et al. Mechanism of inhibition of activated protein C by protein C inhibitor. J Biochem. 1984 Jan;95(1):187–195.
  • Polderdijk SGI, Adams TE, Ivanciu L, et al. Design and characterization of an APC-specific serpin for the treatment of hemophilia. Blood. 2017 Jan 5;129(1):105–113.
  • Zhao XY, Wilmen A, Wang D, et al. Targeted inhibition of activated protein C by a non-active-site inhibitory antibody to treat hemophilia. Nat Commun. 2020 Jun 12;11(1):2992.
  • Prince R, Bologna L, Manetti M, et al. Targeting anticoagulant protein S to improve hemostasis in hemophilia. Blood. 2018 Mar 22;131(12):1360–1371.
  • Favaro P, Finn JD, Siner JI, et al. Safety of liver gene transfer following peripheral intravascular delivery of adeno-associated virus (AAV)-5 and AAV-6 in a large animal model. Hum Gene Ther. 2011 Jul;22(7):843–852.
  • Laffan MA, Rangarajan S, Lester W, et al. Hemostatic results for up to 6 years following treatment with valoctocogene roxaparvovec, an AAV5-hFVIII-SQ gene therapy for severe hemophilia A. Presentation OC 21.2. ISTH Congress, 9-13 Jul, 2022.
  • Fong S, Yates B, Sihn CR, et al. Interindividual variability in transgene mRNA and protein production following adeno-associated virus gene therapy for hemophilia A. Nat Med. 2022 Apr;28(4):789–797.
  • Chapin JC, Ayash-Rashkovsky M, Kenniston J, et al. A translational analysis of immune components in peripheral blood from severe hemophilia A patients treated with TAK-754, an AAV8 vector with a codon-optimized B-domain deleted factor VIII transgene. Presentation PB0211. ISTH Congress, 9-13 Jul, 2022.
  • https://investors.biomarin.com/2022-08-24-First-Gene-Therapy-for-Adults-with-Severe-Hemophilia-A,-BioMarins-ROCTAVIAN-TM-valoctocogene-roxaparvovec-,-Approved-by-European-Commission-EC (Accessed on 2022 Oct 19
  • https://www.fda.gov/media/163466/download. cited 2023 Jan 30.
  • Itzler R, Miller J, Robson R, et al. Improvements in health-related quality of life in adults with severe or moderately severe hemophilia B after receiving etranacogene dezaparvovec gene therapy. Presentation OC 01.02. ISTH Congress, 9-13 July, 2022.
  • Pipe Steven W, W.G Leebek Frank, Recht Michael, et al. 2141 Adults with Severe or Moderately Severe Hemophilia B Receiving Etranacogene Dezaparvovec in the HOPE-B Phase 3 Clinical Trial Continue to Experience a Stable Increase in Mean Factor IX Activity Levels and Durable Hemostatic Protection after 24 Months’ Follow-up. [abstract]. ASH. 2022; 801;1. https://ash.confex.com/ash/2022/webprogram/Paper166135.html cited 2023 Jan 27.
  • 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 Jan 2;382(1):29–40.
  • https://hemophilianewstoday.com/2020/12/11/high-dose-gene-therapy-sb-525-fviii-levels-prevents-bleeds-alta-trial-data/ (Accessed on 2022 Nov 17
  • Young G, Chowdary P, Barton S, et al. Results from B-LIEVE, a phase 1/2 dose confirmation study of FLT180a AAV gene therapy in patients with hemophilia B. Presentation PB0213. ISTH Congress, 9-13 Jul, 2022.
  • Lawton S, Manson M, Fan M, et al. Liver sinusoidal endothelial cells targeted with ultrasound mediated gene delivery shows long-term FVIII expression in hemophilia A mice. Presentation OC 12.5. ISTH Congress, 9-13 Jul, 2022.
  • Wang X, Shin SC, Chiang AFJ, et al. Intraosseous delivery of lentiviral vectors targeting factor VIII expression in platelets corrects murine hemophilia A. Mol Ther. 2015 Apr;23(4):617–626.
  • Chen C, Cai X, Miao C, et al. Rescue of the endogenous FVIII expression in hemophilia A mice using CRISPR/ Cas9 mRNA LNPs. Presentation OC 12.3. ISTH Congress, 9-13 Jul, 2022.
  • Zintner SM, Small JC, Pavani G, et al. Gene-based FVIIa prophylaxis modulates the spontaneous bleeding phenotype of hemophilia A rats. Blood Adv. 2019 Feb 12;3(3):301–311.
  • Barbon E, Ayme G, Mohamadi A, et al. Single-domain antibodies targeting antithrombin reduce bleeding in hemophilic mice with or without inhibitors. EMBO Mol Med. 2020 Apr 7;12(4):e11298.
  • Shi Q, Wilcox DA, Fahs SA, et al. Lentivirus-mediated platelet-derived factor VIII gene therapy in murine haemophilia A. J Thromb Haemost. 2007 Feb;5(2):352–361.
  • Du LM, Nurden P, Nurden AT, et al. Platelet-targeted gene therapy with human factor VIII establishes haemostasis in dogs with haemophilia A. Nat Commun. 2013;4:2773.
  • Accessed on 2022 Sept 25. https://www.hemophilia.org/news/sigilon-announces-a-priority-shift-in-light-of-clinical-trial-setbacks