Abstract
In hemophilia, the unmet needs regarding adherence to prophylaxis and lack of effective long-term prophylaxis regimens, especially in patients with inhibitors, led to the production of emicizumab, the first non-factor medicine for subcutaneous administration in patients with severe and moderate hemophilia A with or without factor VIII inhibitors. This review describes the research steps behind the development of this game-changing medication as well as its success in the prophylaxis of bleeding episodes, as witnessed by the results of pivotal clinical trials but also by real-life use in the frame of a still expanding global market. We also discuss potential and actual adverse events and the nuances related to clinical use, such as laboratory monitoring, development of neutralizing antidrug antibodies, risk of thrombosis/hypercoagulability and role in the management of surgical operations. The potential of emicizumab to prevent bleeding in other congenital and acquired coagulation disorders is also outlined.
Introduction
The history of hemophilia A (HA) treatment has been marked by solid innovation and progress, intended to improve the quality of life of individuals with this bleeding disorder. Emicizumab has contributed in a major way to this revolution as the first of the non-clotting factor options currently available to prevent hemorrhagic events in patients with congenital and acquired HA, with and without alloantibody inhibitors.
Prophylaxis strategies to prevent bleeding are considered the standard of care in HA.1 The benefits of prophylaxis were confirmed by the sentinel randomized, prospective, multicenter trials by Lessinger et al. for patients with inhibitors and by Manco-Johnson et al. for inhibitor-free patients.2,3 Both studies demonstrated that, compared to on-demand therapy, prophylaxis with infusions of anti-inhibitor complex bypassing activity concentrates, or of recombinant factor VIII (rFVIII), prevents joint damage and decreases the frequency of bleeding in joints and other sites in individuals with severe HA with or without inhibitors. The main impediments to success of such regimens have been adherence to the demands of predominantly twice weekly intravenous protocols for factor administration and the availability over time of functional venous accesses for the infusions. With the development and introduction of the recombinant, humanized, bispecific FVIII-mimetic procoagulant antibody, emicizumab, into the therapeutic armamentarium, the subcutaneous administration of hemostatic agents has improved adherence with equivalent safety and efficacy profiles compared to recombinant and plasma-derived concentrates for HA with or without inhibitors.
This review describes the history and pharmacology behind the development of emicizumab, the drug’s initial success for prophylaxis against bleeds associated with alloantibodies neutralizing FVIII, its subsequent usefulness in prophylaxis regimens to prevent bleeds due to HA without inhibitors, and its potential use in other bleeding disorders. The review also discusses complications and the nuances associated with the use of emicizumab, such as laboratory monitoring, development of specific neutralizing antidrug antibodies, and thrombotic/hypercoagulability potential, and shows why this game-changing medication continues to capture an ever-expanding global market share among the hemostatic agents commercially available for the prevention of hemorrhagic episodes in subjects with HA.
Development
The evolution of emicizumab as a therapeutic procoagulant reflects the convergence of intellectual insight into the molecular mechanisms of thrombin generation mediated through activated factor VIII (FVIIIa) with the prowess of the pharmaceutical industry to innovate and bioengineer a molecular “linker”. The concept of binding activated factor IX (FIXa) to factor X (FX) via a bispecific antibody to function as the FVIII cofactor was ascribed by Kitazawa and Shima to Dr. Kunihiro Hattori.4 He and his research group at Chugai Pharmaceutical Co., Ltd. (Chugai) appreciated that the distance between the FIXa-and FX-binding sites of FVIIIa on a phospholipid template would be similar to that between the two antigen-binding sites of human IgG. Thus began the era of FVIII-mimetic therapy. Translating the concept of FVIII mimetics into a practical biopharmaceutical was a monumental and momentous task,5-9 summarized in Figure 1.
Dosing
Using population pharmacokinetic modeling and model-based simulations derived from phase I and II trials, the minimal emicizumab plasma concentrations expected to achieve zero bleeds over a 1-year period in at least 50% of patients were determined10 and applied to formulate the dosing regimens employed in the pivotal phase III HAVEN trials: a subcutaneous loading dose of 3 mg/kg once weekly for a total of 4 weeks and three different subcutaneous maintenance doses starting at week 5: these doses were 1.5 mg/kg once every week, 3 mg/kg once every 2 weeks and 6 mg/kg once every 4 weeks. All regimens have shown comparable efficacy and similar side-effect profiles.
Pivotal clinical trials: establishing efficacy and safety
In the era of very effective FVIII replacement therapies resulting in a median annualized bleeding rate close to zero in most individuals with severe HA, the development of neutralizing anti-FVIII inhibitors has emerged as the highest priority complication of repeated exposure to FVIII products. This immune response to exogenous FVIII occurs in an estimated 20-40% of individuals with severe HA and 3-13% of those with mild-to-moderate HA,11,12 and is associated with increased cost of care, mortality and morbidity, including a higher rate of bleeding, more disability and decreased quality of life.13 Historically, the treatment and prevention of bleeding episodes in patients with FVIII inhibitors was achieved with administration of bypassing agents such as activated prothrombin complex concentrates (aPCC) and recombinant activated factor VII (rFVIIa). In practice, these products provided adequate hemostatic efficacy, although this was temporary and cumbersome to maintain, and less effective than FVIII replacement in patients without inhibitors.14
Thus, the promise of emicizumab as a convenient-to-administer, subcutaneous, non-factor FVIIIa mimetic with durable effectiveness was greatly anticipated when the HAVEN 115 and 216 clinical trials were launched. These phase III studies assessed the efficacy, safety, and pharmacokinetics of once-weekly subcutaneous emicizumab prophylaxis in patients with severe HA and FVIII inhibitors and led to the approval of emicizumab-kxwh by the US Food and Drug Administration (FDA) on November 16, 2017. Subsequent studies were designed to broaden the scope of emicizumab use, including patients without inhibitors (HAVEN 3 and 4),17,18 patients of the Asia-Pacific region (HAVEN 5),19 those with non-severe HA (HAVEN 6)20 and infants (HAVEN 7).21 Different dosing regimens were evaluated and showed the versatility of the drug. These results led to the US FDA and European Medicines Agency (EMA) approvals in 2018 of the multiple prescribing options for the use of emicizumab for prophylaxis in individuals with HA with or without FVIII inhibitors. Results are summarized in Table 1.
The positive results from these clinical trials, as well as the convenient dosing and administration regimens, have led to a rapid uptake for emicizumab in the international HA treatment marketplace. Since 2018, emicizumab is being used by over 12,500 patients22 and generated $3.6 billion in sales in 2022 as the 34th best-selling pharmaceutical of 2022.23 Its availability in developing nations is also well-established following distribution of 2,528,730 mg by the World Federation of Haemophilia to 806 patients in Africa, Latin America, Asia, and Eastern Europe. Of these individuals, 55% were under 12 years old and 64% were non-inhibitor patients.22
Practical therapeutic concerns and questions
Laboratory testing
As the use of emicizumab has increased, so too has the desire to have a routine laboratory assay available to monitor safety and efficacy. This would allow for individualization of dosing and lower costs. However, emicizumab interferes with clotting-based assays, while assays using immunological or chromogenic techniques are not affected24,25 (Table 2).
Global coagulation testing techniques have been proposed to estimate the hemostatic potential of emicizumab. Thrombin generation assays and thromboelastography have been the most used platforms in this area.26-28 Although these assays may reflect changes from baseline coagulation potential following emicizumab, they do not provide “true FVIII equivalence” in vivo and, in fact, a thrombin generation assay may overestimate thrombin levels in plasma specimens in the presence of emicizumab, since the assay is not modulated by the presence of naturally occurring circulating thrombin inhibitors, such as α2 macroglobulin.28 Thus, thrombin-generation assays may lead to overconfidence and faulty clinical decision-making regarding the hemostatic response to emicizumab. It has been suggested that thrombin generation assays may be best used to limit the occurrence of adverse thromboembolic events when other procoagulant medications are administered to treat breakthrough bleeds in individuals on maintenance emicizumab regimens.26 This was poignantly apparent in the HAVEN 1 study, in swhich five patients experienced thromboembolic events when emicizumab was combined with multiple high doses of aPCC. In vitro plasma spiking studies corroborated the marked synergistic effects on thrombin generation assays when aPCC was added to emicizumab.29 However, despite suggestions that there is a direct correlation between the clinical bleeding phenotype of patients with hemophilia and their thrombin generation capacity, real-world experience with emicizumab has challenged this in sporadic clinical instances.30
Would measurement of emicizumab concentrations in plasma provide a more accurate FVIII equivalence? Extensive in vitro studies have not been able to establish true FVIII equivalence for emicizumab, probably due to the different modes of action to activate FX and to the vagaries in the methods of the biochemical assays.28 Kitazawa et al. approached this issue by developing a primate bleeding model of acquired HA.31 Using rFVIII as the FVIII calibrator, the therapeutic emicizumab level of about 55 mg/mL had an estimated FVIII equivalence activity level of 10-20%.28 The HAVEN 1 study demonstrated a steady-state mean emicizumab level of 55 mg/mL (range, 35-70 mg/mL). Even though emicizumab assays are becoming more available commercially, it is anticipated that their use will be somewhat limited, perhaps to confirm a patient’s adherence to prophylaxis and for confirmation of the presence of neutralizing anti-emicizumab antibodies.
Adverse events associated with emicizumab administration
Injection site reactions/hypersensitivity
Thus far, the administration of emicizumab has been remarkably routine and uneventful. Gelbenegger et al. summarized the adverse events recorded in all the HAVEN clinical trials.32 The most frequently observed adverse events were local injection site reactions (erythema, pain, and pruritus), which were reported in 26.1% of treated participants. The reactions were mild to moderate in severity and resolved without treatment in over 90% of cases. Anaphylaxis was extremely rare. There were two adult cases of rhabdomyolysis, which occurred following intense physical exercise.
Thromboembolic complications/thrombotic microangiopathy
The most concerning of the adverse events reported with emicizumab during clinical trials has been the occurrence of thromboembolic events and thrombotic microangiopathy, as summarized in Table 1. Critically, these complications were associated with concomitant replacement therapy with aPCC at cumulative doses over 100 U/kg/day for more than 1 day. An updated summary of thromboembolic events documented 37 additional cases of non-aPCC-related events accumulated from post-marketing reports and registries as of May, 2021.33 Of these, 92% occurred in patients with pre-existing cardiovascular or thrombotic risk factors, including age over 50; 17 (45.9%) occurred in patients with inhibitors, and seven (18.9%) were associated with thrombotic occlusion of a central venous access device. Only six of the 30 individuals with non-thromboembolic events not related to a central venous access device discontinued their emicizumab prophylaxis. There were four deaths in this cohort: two patients with myocardial infarction and two elderly individuals over 70 years old with pneumonia and disseminated intravascular coagulation. Robust real-world and long-term experience with emicizumab is summarized in Table 3.34-39 No pediatric participants have experienced thromboembolic events in the HAVEN trials and no pediatric deaths have been reported in the HAVEN series. In contrast, the real-world use of emicizumab has been associated with the death of an Israeli child, who died after developing a central venous access device-related thrombosis.30 Undoubtedly, with the rapid worldwide uptake of emicizumab prophylaxis regimens in HA, additional reports of thrombotic morbidity and mortality will emerge. The use of registries will be critical to monitor the safety of this and other non-factor replacement strategies. It also highlights the fact that emicizumab may not be the optimal treatment strategy for all individuals with HA with or without inhibitors and that careful selection of patients will be necessary to achieve favorable risk/benefit outcomes. The black box warnings required by the FDA in the drug’s package insert serve as a reminder that the use of emicizumab may carry both known and unanticipated risks.
Antidrug antibodies
Emicizumab was associated with the development of antidrug antibodies in 5.1% of patients (34/668) who participated in the HAVEN 1-4 clinical trials.40 Most patients did not experience a change in emicizumab plasma concentrations or an increase in bleeding events; however, neutralizing antibodies were detected in <1% of cases and were presumed to be responsible for decreasing the plasma concentration of emicizumab and its subsequent loss of efficacy. The analysis of these antidrug antibodies indicate that there are at least two mechanisms by which they can reduce emicizumab’s efficacy: by inhibiting the direct binding of emicizumab to its targets on either the FXa or FX Fab regions or by binding to the Fc region of emicizumab and thus increasing its clearance from plasma.41,42 It should be noted that antidrug antibodies do not affect the efficacy of FVIII replacement or bypassing therapies to prevent or reverse hemorrhagic events. The development of an anti-emicizumab antibody should be suspected when a patient experiences an increased frequency of breakthrough bleeds after having demonstrated a previously low annualized bleeding rate with emicizumab. Confirmation of neutralizing antidrug antibodies can be approached simply by demonstrating a newly prolonged aPTT and lowered one-stage FVIII or chromogenic FVIII activity with human reagents, since the aPTT should be normal and the FVIII activities high in emicizumab-treated patients. FVIII activity determined chromogenically with bovine substrate will also be very decreased.
Breakthrough bleeding
In the pooled analysis of HAVEN 1-4 trials, there were rare breakthrough bleeds, either spontaneous or traumatic, while on emicizumab prophylaxis regimens. Over 90% of the participants had no recorded breakthrough target joint hemarthroses during up to 144 weeks of emicizumab administration. Thus, the small number of breakthrough bleeding events and improved bleeding control overall led to reduced joint morbidity; the longer the emicizumab was used, the healthier the joint became.40 This concept was challenged by an Israeli cohort included in Table 3, in which all spontaneous bleeds occurred in previous target joints. Perhaps a hint into the mechanism of spontaneous breakthrough bleeds despite adequate hemostatic levels of emicizumab and in the absence of antidrug antibodies lies in the integrity of the fibrin clot which forms after thrombin is generated in plasma by emicizumab. Shimonishi et al. analyzed the structure of fibrin clots induced by a prothrombin/aPTT trigger reagent in the presence of emicizumab (50 mg/mL, i.e., the clinically therapeutic concentration) and showed that the fibrin structure was comparable to that of ~12% FVIII.43 Similarly, fibrin clots generated in the presence of 100 mg/mL emicizumab had a structure equivalent to 20-30% FVIII. These in vitro studies indicate improved clot structure with emicizumab in HA plasma but do not provide information regarding clot stability. Fibrin clots formed in HA plasma have been demonstrated to be more fragile and weaker compared to normal control fibrin clots and appear to be more susceptible to fibrinolysis. Thus, the FVIII activity equivalence conveyed by therapeutic plasma levels of emicizumab may not accurately reflect hemostatic adequacy in all clinical scenarios.
The preferred treatments for breakthrough bleeding include infusions of rFVIIa concentrates to bypass allogeneic FVIII neutralizing antibody inhibitors and of rFVIII concentrates in the absence of FVIII inhibitors. The success of the latter strategy is based on the premise that the binding affinities of exogenously administered FVIII for both FIXa and FX are considerably higher than those of emicizumab (low-to-high nanomolar range vs. micromolar range), thus allowing for a competitive advantage to FVIII for FXa generation. Furthermore, FVIIIa activity in the tenase complex is modulated by circulating natural anticoagulants, i.e., activated protein C, and by spontaneous dissociation of the A2 domain, thus limiting the degree of FXa generation. This helps to prevent the development of hypercoagulability.44
The use of aPCC can be considered as a less optimal alternative to treat breakthrough bleeds in the presence of a FVIII inhibitor, and extreme caution is necessary to avoid creating a hypercoagulable state; dose modulation is critical (see previous discussion).
Uncertain treatment scenarios, strategies, and benefits
Emicizumab in males over 50 years old
The safety of emicizumab in older males with severe HA with or without allogeneic FVIII antibody inhibitors has been a topic of concern because the drug’s mechanism of action is to maintain a continuous in vivo procoagulant environment.
Elderly individuals with severe HA were significantly under-represented in the HAVEN 1, 3, 4, and STASEY clinical trials. Of a total of 504 patients in the overall study population, 70 were aged 50 to 64 years and 22 were ≥65 years. There was no difference in efficacy or safety in elderly individuals compared to the younger participants in the clinical trials. The real-world experience with emicizumab therapy in individuals over 50 years of age is somewhat limited but growing rapidly. An Israeli cohort of patients older than 50 years showed the efficacy of emicizumab, with 41% of elderly individuals experiencing an annualized bleeding rate of zero, but safety concerns remain39 (Table 3).
Emicizumab in previously untreated patients and for immune tolerance induction strategies
The National Hemophilia Foundation of the USA and the World Federation of Haemophilia have both endorsed emicizumab as an important therapeutic option to achieve primary prophylaxis in infants with severe HA. It is recommended that primary prophylaxis is initiated at any time after birth (≤2 years old or even before the first clinical bleed), given the increased risks of developing potentially life-threatening spontaneous and traumatic intracranial hemorrhages without the initiation of primary prophylaxis. Accumulating data from the HAVEN 7 clinical trial and single institution experience have generated data to support the prophylactic efficacy and safety of emicizumab in infants. Results have shown a high percentage of zero treated bleeds and no neutralizing antidrug antibodies have been observed thus far. In addition, no deaths, no thromboembolic events, no thrombotic microangiopathy and no anti-emicizumab neutralizing antibodies have been reported.21 One infant experienced major bleeding after circumcision while on emicizumab, and this episode was associated with suboptimal levels of thrombin generation despite administration of emicizumab.45 Prospective and longitudinal real-world clinical safety and laboratory data in previously untreated patients are being collected.
The emergence of emicizumab as an easily administered subcutaneous procoagulant has provided a viable alternative to achieve and maintain the benefits of primary prophylaxis while mitigating many of the pitfalls of traditional FVIII replacement. Practically, primary prophylaxis using exogenous intravenous FVIII replacement at least two or three times weekly is delayed until the child is active and walking at around 12-15 months of age (ideally before the first bleed but most commonly after the first few bleeds have occurred) when traumatic bleeds are most likely to prevail, and venous access is more available. However, this approach has been associated with the development of allogeneic FVIII neutralizing inhibitory antibodies in at least 30% of previously untreated patients on primary prophylaxis.46 FVIII inhibitors develop very rarely after the patient reaches 50 exposure days of exogenous FVIII administration. Venous access challenges, maintaining adherence to the prophylaxis protocol over time, and the financial and personal burdens of family caregivers are all additional potential potent impediments to the success of primary prophylaxis.
Primary prophylaxis based on emicizumab alone can theoretically be initiated before the first bleed and simplify the prophylaxis process in previously untreated patients. However, some treaters worry that this strategy would simply delay the emergence of FVIII inhibitors and may complicate future surgery or other bleeding scenarios in which FVIII administration will be necessary. Clinical trials are underway to compare the incidence of inhibitor development in children who receive emicizumab alone versus combined with concurrent FVIII replacement.
Emicizumab prophylaxis is also being studied as a component of immune tolerance induction regimens to determine the rate of inhibitor recurrence if concurrent FVIII replacement is discontinued after immune tolerance has been successfully induced. The reader is referred to a very thoughtful and comprehensive discussion of the debate on immune tolerance induction and treatment decision-making pertinent to previously untreated or minimally treated patients in the era of emicizumab.47
Emicizumab for surgery
Patients who were planned to undergo surgery were excluded from the pivotal clinical trials (HAVEN 1-4), and perioperative management was at the discretion of the individual investigators. There was no specific guidance (per protocol) on surgical management provided by the sponsors and even after regulatory approval of emicizumab by the FDA and EMA, package inserts have provided no re-commendations for how to achieve, monitor, and maintain adequate hemostasis and FVIII/bypass product replacement during surgical procedures. (https://www. hemlibra-hcp.com) Recently, the manufacturer issued a physician/healthcare procedure (https://www.emicizumabinfo.com) which summarizes the most relevant published surgical experiences with emicizumab.
It is crucial to remember that emicizumab prophylaxis provides for incrementally increased hemostasis, which is equivalent to modifying a bleeding phenotype from severe to mild HA. However, emicizumab does not completely normalize hemostasis. Thus, for major surgery it is prudent in the context of a pre-existing emicizumab prophylaxis regimen to provide adjunctive procoagulant therapy with either exogenous FVIII (in the absence of an inhibitor) or with a bypassing agent (aPCC or rFVIIa) in the presence of a neutralizing allogeneic FVIII antibody inhibitor. Doses of aPCC should not exceed 50 U/kg body weight due to the potential development of thrombosis or thrombotic microangiopathy. If prolonged perioperative and postoperative treatment with aPCC is anticipated to be necessary following elective major surgery, it may be necessary to discontinue emicizumab several months prior to the operation (the elimination half-life of emicizumab is approximately 30 days). For minor surgical interventions in the context of emicizumab prophylaxis, additional procoagulant therapies may not be necessary to achieve or maintain adequate hemostasis, but patients should be closely monitored for clinical bleeding.48 These recommendations are based on evidence from clinical trials and real-world experience, summarized in Table 4.49-52 Standardized management as proposed by Castaman et al. would be ideal, but the optimal strategies remain to be determined. Most treaters agree that aPCC should be avoided in surgical scenarios to reduce thromboembolic events or thrombotic microangiopathic complications.
Emicizumab in acquired hemophilia A
Even though acquired HA is a rare bleeding disorder (1 cases per 1-3 million population), the high mortality from hemorrhagic complications which occur in patients with this condition emphasizes the importance of effective treatments for bleeding control and for subsequent eradication of the FVIII neutralizing autoantibody by means of immunosuppression. Current treatment options to manage acquired HA include FVIII bypassing agents (rFVIIa and aPCC) and recombinant porcine (rp) FVIII.53
Anecdotal case reports and limited case series have described the use of emicizumab in patients with acquired HA. The usual scenario has involved administration of emicizumab to prevent recurrent bleeding events, following the initial use of rpFVIII, rFVIIa or aPCC to control the acute bleed. Dosing schemes for emicizumab for both loading and maintenance strategies have varied and the timing of the use of adjunctive immunosuppressive therapies to eradicate the inhibitor have not been standardized.54-57
The growing evidence of clinical benefit prompted a prospective, multicenter, single-arm, phase III clinical trial (AGEHA) in Japan to investigate the safety, efficacy, pharmacokinetics, and pharmacodynamics of emicizumab in acquired HA. Emicizumab efficacy was associated with a remarkable reduction in annualized bleeding rate from 66.4 to zero for all major bleeds. Patients received emicizumab 6 mg/kg subcutaneously on day 1, 3 mg/kg on day 2, and then 1.5 mg/kg once weekly from day 8 thereafter until FVIII activity exceeded 50 IU/dL without the need for exogenous coagulation factor products to treat bleeding events. All received immunosuppressive therapy. There was one asymptomatic deep vein thrombosis,58 which was attributed to emicizumab, and which was detected 9-11 days after receiving three doses of FXIII concentrate (Fibrogammin). The patient had not received any bypassing treatments. In addition, another patient developed a non-neutralizing antidrug antibody, which did not affect plasma half-life. No onset of thrombotic microangiopathy was observed.
The optimal dosing regimen and duration of treatment with emicizumab for acquired HA remain to be elucidated. An important aspect to consider is that the long half-life of emicizumab can maintain a therapeutic effect while immunosuppression achieves low levels of inhibitor. On the other hand, once FVIII levels increase, patients might have an increased risk of thrombosis and emicizumab should be discontinued. There are two ongoing, prospective, open-label, clinical trials, being conducted in parallel in the USA (NCT05345197) and Germany (NCT04188639), which will examine the efficacy of prophylactic emicizumab administered on a scheduled basis to prevent bleeds in patients with acquired HA.
Clinical unknowns
Emicizumab and bone health
There is a direct relationship in severe HA (and hemophilia B) between joint and bone health. Severely decreased FVIII (<5% of normal) will predispose the joint to spontaneous and traumatic bleeds, and recurrent bleeds into the joint will cause synovial hypertrophy, cartilage deterioration, bone destruction, motion limitation and pain. Hemophilic arthropathy can be greatly impeded, if not prevented, by maintaining persistently adequate hemostasis with prophylaxis regimens with replacement of FVIII or the use of non-factor replacement strategies.
Another important consequence of severe FVIII deficiency is the development of reduced bone mineral density, which leads to abnormal bone remodeling, gait disturbance, osteopenia, and ultimately osteoporosis. This may be responsible for the increased bone fracture rate observed in individuals with severe hemophilia. Findings from FVIII-deficient mouse models suggest that decreased thrombin generation and unbalanced FVIII-von Willebrand factor complex formation may impede the osteoblastic activity needed for normal bone formation.59,60 There is a current prospective clinical trial (NCT04131036) to examine bone and joint health over a 3-year period in patients with severe HA treated with emicizumab versus clotting factor prophylaxis. Biomarkers and bone density will also be assessed. Data from patients enrolled in the HAVEN 3 trial did not show significant differences in markers of bone remodeling, cartilage degradation or synthesis, questioning the need for FVIII exposure, but longer follow up may be required.61
With this and any other of the potential physiological roles FVIII may have,60 the question arises as to whether individuals with severe HA who are being treated with emicizumab exclusively for its procoagulant properties will benefit incompletely from or be devoid of the long term non-hemostatic properties of FVIII. This may be most critical in the pre-adolescent years.
Additional clinical applications
The unique procoagulant properties and safety profile of emicizumab have led to the desire to determine whether this drug would be effective at reversing or preventing bleeding induced by other coagulopathies. Yada and Nogami have speculated that any bleeding disorder that has a small amount of activated factor IX available to bind to emicizumab in vivo might be amenable to the procoagulant benefits of increased thrombin generation by the drug62 (Figure 2). Potential uses of emicizumab beyond HA are summarized in Table 5.24,63-71
Concluding remarks
The development of extended half-life rFVIII products certainly represented a substantial progress in the treatment of HA. Nevertheless, although improvement in annualized bleeding rate has been achieved with regular injections, only a minority of patients gain absolute bleeding control or zero bleeds,72 highlighting the need for alternative therapies. Emicizumab has revolutionized HA management. Its main advantages include subcutaneous administration and the possibility of up to monthly injections, which can improve adherence. Results from clinical trials and real-world data are encouraging, showing efficacy and achievement of zero bleeds in a substantial proportion of patients. Hence, indications for the use of emicizumab will probably be expanded to patients with moderate HA, infants with severe HA, and other coagulopathies, including von Willebrand disease and acquired HA.
However, many questions remain unanswered. The main concern, based on results from the HAVEN trials, is the risk of thromboembolic events and thrombotic microangiopathy. Yet post-marketing surveillance programs have shown that risk mitigation strategies, including judicious use of other therapies, such as aPCC, are effective.33 Another area of uncertainty is the management of breakthrough bleeding, which a minority of patients experience. Similarly, more data are needed regarding the management of patients undergoing surgical procedures, and regarding the risks of thrombosis and bleeding in elderly individuals using emicizumab concomitantly with antiplatelet and/or anticoagulant agents.
The pursuit of the ideal treatment for HA continues. The success of emicizumab has led to the development of a new generation of FVIII-mimetic bispecific antibodies, NXT007 and Mim8, which are currently in phase I/II studies.73,74 Betting on the benefits of using FVIII, a new extended half-life rFVIII molecule was created by fusing a dimeric Fc fragment and two XTEN polypeptides to B-domain-deleted rFVIII to achieve a longer circulating half-life. In addition, the D’D3 domain of von Willebrand factor (the physiological FVIII binding site), was appended to the fusion FVIII-Fc molecule in order to circumvent the limitations to prolonging plasma half-life extension imposed by FVIII- von Willebrand factor complex formation, as occurs with all previously available commercial FVIII replacement products.75 Efanesoctocog alfa (BIVV001) has been shown to achieve adequate FVIII plasma activity levels with weekly infusions, resulting in a 77% reduction in the mean annualized bleeding rate, resolution of bleeding episodes in most patients, and improvement in pain and quality of life.76 A long-term safety and efficacy evaluation is planned (NCT04644575). Advantages of efanesoctocog alfa include its use to treat breakthrough bleeding episodes and in the perioperative setting with a simplified weekly regimen.
To rebalance hemostasis, new treatment options are being explored. Fitusiran, a small interfering RNA that decreases hepatic synthesis of antithrombin, has shown a significant decrease in annualized bleeding rate with the advantage of monthly subcutaneous administration, making it a promising product.77 Concizumab, a monoclonal anti-tissue factor pathway inhibitor (TFPI) antibody has also shown comparable results, as far as concerns annualized bleeding rate, with once-daily subcutaneous administration.78 However, thromboembolic events seen in the early phases of development of these products remain a source of concern and longer follow-up is needed. Marstacimab is another anti-TFPI monoclonal antibody with the advantage of weekly subcutaneous dosing. It has shown promising results after 1 year of follow-up and is currently in a phase III study.79 A summary of available treatments is shown in Table 6.
Finally, efforts to find a cure for patients with HA continue. Although gene therapy is beyond the scope of this review, it might be a feasible option in the future. Limitations include variable durability of effectiveness, inability to predict the extent of FVIII incremental response, hepatotoxicity, development of antibodies against the vector, availability in children, and the cost of gene therapy acquisition.
With multiple treatment options available, it is an exciting time for the hemophilia field. Patients and providers will face challenging decisions when choosing the optimal therapy, especially considering that head-to-head comparisons between emicizumab and other treatments are unlikely to be done. Emicizumab has certainly been a game-changer, with robust data showing its safety and efficacy, but the game has not ended. If gene therapy is also approved in children, this will probably be the ideal option for children and young adults who are more likely to look for a cure. Extended half-life FVIII products with improved pharma-cokinetics that guarantee better adherence to treatment will continue to be critical in the management of patients with cardiovascular and thromboembolic risk factors, as well as patients with breakthrough bleeds and in the perioperative setting. Replacement products, which reset hemostatic balance by interfering with the inhibitory and modulatory mechanisms of normal coagulation pathways, e.g., antithrombin and TFPI, are bringing further innovation to the field. HA therapies have joined the era of precision medicine.
Footnotes
- Received August 14, 2023
- Accepted October 26, 2023
Correspondence
Disclosures
PEAA has no conflicts of interest to disclose. PMM has received honoraria for lectures at educational symposia from Roche, Takeda and Werfen. CMK has received funds from Bayer, Genentech, Novo Nordisk and Octapharma and is a member of advisory boards for Bayer, CSL, Genentech, Octapharma, Pfizer, Biomarin, Sangamo and Takeda.
Contributions
PEAA, PMM and CMK contributed equally to writing and editing this manuscript.
References
- Srivastava A, Santagostino E, Dougall A. WFH guidelines for the management of hemophilia, 3rd edition. Haemophilia. 2020; 26(Suppl 6):1-158. Google Scholar
- Leissinger C, Gringeri A, Antmen B. Anti-inhibitor coagulant complex prophylaxis in hemophilia with inhibitors. N Engl J Med. 2011; 365(18):1684-1692. Google Scholar
- Manco-Johnson MJ, Abshire TC, Shapiro AD. Prophylaxis versus episodic treatment to prevent joint disease in boys with severe hemophilia. N Engl J Med. 2007; 357(6):535-544. Google Scholar
- Kitazawa T, Shima M. Emicizumab, a humanized bispecific antibody to coagulation factors IXa and X with a factor VIIIacofactor activity. Int J Hematol. 2020; 111(1):20-30. Google Scholar
- Muto A, Yoshihashi K, Takeda M. Anti-factor IXa/X bispecific antibody (ACE910): hemostatic potency against ongoing bleeds in a hemophilia A model and the possibility of routine supplementation. J Thromb Haemost. 2014; 12(2):206-213. Google Scholar
- Muto A, Yoshihashi K, Takeda M. Anti-factor IXa/X bispecific antibody ACE910 prevents joint bleeds in a long-term primate model of acquired hemophilia A. Blood. 2014; 124(20):3165-3171. Google Scholar
- Shima M, Hanabusa H, Taki M. Factor VIII-mimetic function of humanized bispecific antibody in hemophilia A. N Engl J Med. 2016; 374(21):2044-2053. Google Scholar
- Shima M, Hanabusa H, Taki M. Long-term safety and efficacy of emicizumab in a phase 1/2 study in patients with hemophilia A with or without inhibitors. Blood Adv. 2017; 1(22):1891-1899. Google Scholar
- Uchida N, Sambe T, Yoneyama K. 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. Google Scholar
- Yoneyama K, Schmitt C, Kotani N. A pharmacometric approach to substitute for a conventional dose-finding study in rare diseases: example of phase III dose selection for emicizumab in hemophilia A. Clin Pharmacokinet. 2018; 57(9):1123-1134. Google Scholar
- Gouw SC, van der Bom JG, Ljung R. Factor VIII products and inhibitor development in severe hemophilia A. N Engl J Med. 2013; 368(3):231-239. Google Scholar
- Peyvandi F, Mannucci PM, Garagiola I. A randomized trial of factor VIII and neutralizing antibodies in hemophilia A. N Engl J Med. 2016; 374(21):2054-2064. Google Scholar
- Earnshaw SR, Graham CN, McDade CL, Spears JB, Kessler CM. Factor VIII alloantibody inhibitors: cost analysis of immune tolerance induction vs. prophylaxis and on-demand with bypass treatment. Haemophilia. 2015; 21(3):310-319. Google Scholar
- DiMichele DM, Hoots WK, Pipe SW, Rivard GE, Santagostino E. International workshop on immune tolerance induction: consensus recommendations. Haemophilia. 2007; 13(Suppl 1):1-22. Google Scholar
- Oldenburg J, Mahlangu JN, Kim B. Emicizumab prophylaxis in hemophilia A with inhibitors. N Engl J Med. 2017; 377(9):809-818. Google Scholar
- Young G, Liesner R, Chang T. A multicenter, open-label phase 3 study of emicizumab prophylaxis in children with hemophilia A with inhibitors. Blood. 2019; 134(24):2127-2138. Google Scholar
- Mahlangu J, Oldenburg J, Paz-Priel I. Emicizumab prophylaxis in patients who have hemophilia A without inhibitors. N Engl J Med. 2018; 379(9):811-822. Google Scholar
- Pipe SW, Shima M, Lehle M. 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(6):e295-e305. Google Scholar
- Yang R, Wang S, Wang X. Prophylactic emicizumab for hemophilia A in the Asia-Pacific region: a randomized study (HAVEN 5). Res Pract Thromb Haemost. 2022; 6(2):e12670. Google Scholar
- Negrier C, Mahlangu J, Lehle M. Emicizumab in people with moderate or mild haemophilia A (HAVEN 6): a multicentre, open-label, single-arm, phase 3 study. Lancet Haematol. 2023; 10(3):e168-e177. Google Scholar
- Pipe SW, Collins P, Dhalluin C. Emicizumab prophylaxis for the treatment of infants with severe hemophilia A without factor VIII inhibitors: results from the interim analysis of the HAVEN 7 study. Blood. 2022; 140(Suppl 1):457-459. Google Scholar
- Mahlangu J, Iorio A, Kenet G. Emicizumab state-of-the-art update. Haemophilia. 2022; 28(Suppl 4):103-110. Google Scholar
- Buntz B. The 50 best-selling pharmaceuticals of 2022: COVID-19 vaccines poised to take a step back. 2023. Publisher Full TextGoogle Scholar
- Adamkewicz JI, Chen DC, Paz-Priel I. Effects and interferences of emicizumab, a humanised bispecific antibody mimicking activated factor VIII cofactor function, on coagulation assays. Thromb Haemost. 2019; 119(7):1084-1093. Google Scholar
- Adamkewicz JI, Kiialainen A, Paz-Priel I. Effects and interferences of emicizumab, a humanized bispecific antibody mimicking activated factor VIII cofactor function, on lupus anticoagulant assays. Int J Lab Hematol. 2020; 42(2):e71-e75. Google Scholar
- Dargaud Y, Lienhart A, Janbain M. Use of thrombin generation assay to personalize treatment of breakthrough bleeds in a patient with hemophilia and inhibitors receiving prophylaxis with emicizumab. Haematologica. 2018; 103(4):e181-e183. Google Scholar
- Kizilocak H, Yukhtman CL, Marquez-Casas E. Management of perioperative hemostasis in a severe hemophilia A patient with inhibitors on emicizumab using global hemostasis assays. Ther Adv Hematol. 2019; 10:2040620719860025. Google Scholar
- Lenting PJ. Laboratory monitoring of hemophilia A treatments: new challenges. Blood Adv. 2020; 4(9):2111-2118. Google Scholar
- Hartmann R, Feenstra T, Valentino L, Dockal M, Scheiflinger F. In vitro studies show synergistic effects of a procoagulant bispecific antibody and bypassing agents. J Thromb Haemost. 2018; 16(8):1580-1591. Google Scholar
- Barg AA, Budnik I, Avishai E. Emicizumab prophylaxis: prospective longitudinal real-world follow-up and monitoring. Haemophilia. 2021; 27(3):383-391. Google Scholar
- Kitazawa T, Igawa T, Sampei Z. 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. Google Scholar
- Gelbenegger G, Schoergenhofer C, Knoebl P, Jilma B. Bridging the missing link with emicizumab: a bispecific antibody for treatment of hemophilia A. Thromb Haemost. 2020; 120(10):1357-1370. Google Scholar
- Howard M, McKinley D, Sanabria F, Ko RH, Nissen F. Evaluation of the safety of emicizumab prophylaxis in persons with hemophilia A: an updated summary of thrombotic events and thrombotic microangiopathies. Blood. 2021; 138(Suppl 1):3186. Google Scholar
- Hassan E, Jonathan L, Jayashree M. Real-world experience on the tolerability and safety of emicizumab prophylaxis in paediatric patients with severe haemophilia A with and without FVIII inhibitors. Haemophilia. 2021; 27(6):e698-e703. Google Scholar
- Ebbert PT, Xavier F, Seaman CD, Ragni MV. Emicizumab prophylaxis in patients with haemophilia A with and without inhibitors. Haemophilia. 2020; 26(1):41-46. Google Scholar
- Buckner T, Daoud N, Croteau SE. ATHN 7: a natural history cohort study of the safety, effectiveness, and practice of treatment for people with hemophilia-demographics and preliminary results [abstract]. Res Pract Thromb Haemost. 2023. Publisher Full TextGoogle Scholar
- Nissen F, Jiang Y, Hiew HJ. Real-world safety of emicizumab: interim analysis of the European Haemophilia Safety Surveillance (EUHASS) database. Blood. 2022; 140(Suppl 1):469-470. Google Scholar
- Levy-Mendelovich S, Brutman-Barazani T, Budnik I. Real-world data on bleeding patterns of hemophilia A patients treated with emicizumab. J Clin Med. 2021; 10(19):4303. Google Scholar
- Misgav M, Brutman-Barazani T, Budnik I. Emicizumab prophylaxis in haemophilia patients older than 50 years with cardiovascular risk factors: real-world data. Haemophilia. 2021; 27(2):253-260. Google Scholar
- Callaghan MU, Negrier C, Paz-Priel I. 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. Google Scholar
- Kaneda M, Kawasaki R, Matsumoto N. Detailed analysis of anti-emicizumab antibody decreasing drug efficacy, using plasma samples from a patient with hemophilia A. J Thromb Haemost. 2021; 19(12):2938-2946. Google Scholar
- Valsecchi C, Gobbi M, Beeg M. Characterization of the neutralizing anti-emicizumab antibody in a patient with hemophilia A and inhibitor. J Thromb Haemost. 2021; 19(3):711-718. Google Scholar
- Shimonishi N, Nogami K, Ogiwara K. Emicizumab improves the stability and structure of fibrin clot derived from factor VIII-deficient plasma, similar to the addition of factor VIII. Haemophilia. 2020; 26(3):e97-e105. Google Scholar
- Lenting PJ, Denis CV, Christophe OD. Emicizumab, a bispecific antibody recognizing coagulation factors IX and X: how does it actually compare to factor VIII?. Blood. 2017; 130(23):2463-2468. Google Scholar
- Barg AA, Avishai E, Budnik I. Emicizumab prophylaxis among infants and toddlers with severe hemophilia A and inhibitors-a single-center cohort. Pediatr Blood Cancer. 2019; 66(11):e27886. Google Scholar
- Kempton CL, Meeks SL. Toward optimal therapy for inhibitors in hemophilia. Blood. 2014; 124(23):3365-3372. Google Scholar
- Glonnegger H, Andresen F, Kapp F. Emicizumab in children: bleeding episodes and outcome before and after transition to emicizumab. BMC Pediatr. 2022; 22(1):487. Google Scholar
- Wieland I. Emicizumab for all pediatric patients with severe hemophilia A. Hamostaseologie. 2022; 42(2):104-115. Google Scholar
- Santagostino E, Oldenburg J, Chang T. Surgical experience from four phase III studies (HAVEN 1-4) of emicizumab in persons with haemophilia A (PwHA) with or without FVIII inhibitors abstract. 2023. Publisher Full TextGoogle Scholar
- Castaman G, Windyga J, Alzahrani H. Surgical experience from the phase III STASEY trial of emicizumab prophylaxis in persons with hemophilia A with FVIII inhibitors: final analysis. Blood. 2021; 138(Suppl 1):344. Google Scholar
- Barg AA, Livnat T, Budnik I. Emicizumab treatment and monitoring in a paediatric cohort: real-world data. Br J Haematol. 2020; 191(2):282-290. Google Scholar
- Castaman G, Linari S, Pieri L. Safe and successful surgical outcome in persons with hemophilia A with and without inhibitors treated with emicizumab: a large, single center, real-world experience. J Clin Med. 2023; 12(6):2317. Google Scholar
- Kruse-Jarres R, Kempton CL, Baudo F. Acquired hemophilia A: updated review of evidence and treatment guidance. Am J Hematol. 2017; 92(7):695-705. Google Scholar
- Hansenne A, Hermans C. Emicizumab in acquired haemophilia A: about two clinical cases and literature review. Ther Adv Hematol. 2021; 12:20406207211038193. Google Scholar
- Knoebl P, Thaler J, Jilma P. Emicizumab for the treatment of acquired hemophilia A. Blood. 2021; 137(3):410-419. Google Scholar
- Pezeshkpoor B, Sereda N, Berkemeier A. Neutralizing anti-emicizumab antibodies in a patient with acquired hemophilia A [abstract]. 2023. Publisher Full TextGoogle Scholar
- Poston JN, Al-Banaa K, von Drygalski A. Emicizumab for the treatment of acquired hemophilia A: a multicenter US case series. Blood. 2021; 138(Suppl 1):496. Google Scholar
- Shima M, Amano K, Ogawa Y. A prospective, multicenter, open-label phase III study of emicizumab prophylaxis in patients with acquired hemophilia A. J Thromb Haemost. 2023; 21(3):534-545. Google Scholar
- Baud’huin M, Duplomb L, Teletchea S. Factor VIII-von Willebrand factor complex inhibits osteoclastogenesis and controls cell survival. J Biol Chem. 2009; 284(46):31704-31713. Google Scholar
- Samuelson Bannow B, Recht M, Negrier C. Factor VIII: long-established role in haemophilia A and emerging evidence beyond haemostasis. Blood Rev. 2019; 35:43-50. Google Scholar
- Kiialainen A, Niggli M, Kempton CL. Effect of emicizumab prophylaxis on bone and joint health markers in people with haemophilia A without factor VIII inhibitors in the HAVEN 3 study. Haemophilia. 2022; 28(6):1033-1043. Google Scholar
- Yada K, Nogami K. Novel insights and new developments regarding coagulation revealed by studies of the anti-factor IXa (activated factor IX)/factor X bispecific antibody, emicizumab. Arterioscler Thromb Vasc Biol. 2020; 40(5):1148-1154. Google Scholar
- Ogiwara K, Minami H, Nogami K. Anti-FIXa/FX bispecific antibody (emicizumab) enhances plasma procoagulant activity in hemophilia B in the presence of very low level of factor IX. Res Pract Thromb Haemost: Res Pract Thromb Haemost. 2017; 1(Suppl 1):1438-1451. Google Scholar
- Chau J, Sternberg A, Pishko A. Improved procoagulant activity of hemophilia B causing dysfunctional factor IX variants with factor VIII mimetics [abstract]. 2023. Publisher Full TextGoogle Scholar
- Minami H, Nogami K, Yada K. Emicizumab, the bispecific antibody to factors IX/IXa and X/Xa, potentiates coagulation function in factor XI-deficient plasma in vitro. J Thromb Haemost. 2019; 17(1):126-137. Google Scholar
- Barg AA, Avishai E, Budnik I. The potential role of emicizumab prophylaxis in severe von Willebrand disease. Blood Cells Mol Dis. 2021; 87:102530. Google Scholar
- Barg AA, Kenet G, Livnat T. The potential role of emicizuamb prophylaxis in severe von Willebrand disease. Blood. 2020; 136(Suppl 1):20. Google Scholar
- Shanmukhaiah C, Jijina F, Kannan S. Efficacy of emicizumab in von Willebrand disease (VWD) patients with and without alloantibodies to von Willebrand factor (VWF): report of two cases and review of literature. Haemophilia. 2022; 28(2):286-291. Google Scholar
- Thomas VM, Abou-Ismail MY, Lim MY. Off-label use of emicizumab in persons with acquired haemophilia A and von Willebrand disease: a scoping review of the literature. Haemophilia. 2022; 28(1):4-17. Google Scholar
- Yaoi H, Shida Y, Kitazawa T, Shima M, Nogami K. Activated factor VIII-mimicking effect by emicizumab on thrombus formation in type 2N von Willebrand disease under high shear flow conditions. Thromb Res. 2021; 198:7-16. Google Scholar
- Tripodi A, Chantarangkul V, Padovan L. Effect of emicizumab on global coagulation assays for plasma supplemented with apixaban or argatroban. J Thromb Thrombolysis. 2020; 49(3):413-419. Google Scholar
- Mannucci PM. Recombinant FVIII: the milestone of modern hemophilia treatment. Haematologica. 2023; 108(5):1201-1202. Google Scholar
- Kjellev SL, Østergaard H, Greisen PJ. Mim8 - a next-generation FVIII mimetic bi-specific antibody - potently restores the hemostatic capacity in hemophilia A settings in vitro and in vivo. Blood. 2019; 134(Suppl_1):96. Google Scholar
- Yamaguchi K, Soeda T, Sato M. Pharmacology and pharmacokinetics of NXT007; emicizumab-based engineered FIXa/ FX bispecific antibody with improved properties. Blood. 2020; 136(Suppl 1):19. Google Scholar
- Konkle BA, Shapiro AD, Quon DV. BIVV001 fusion protein as factor VIII replacement therapy for hemophilia A. N Engl J Med. 2020; 383(11):1018-1027. Google Scholar
- von Drygalski A, Chowdary P, Kulkarni R. Efanesoctocog alfa prophylaxis for patients with severe hemophilia A. N Engl J Med. 2023; 388(4):310-318. Google Scholar
- Kenet G, Nolan B, Zulfikar B. A phase 3 study (ATLAS-PPX) to evaluate efficacy and safety of fitusiran, an siRNA therapeutic, in people with haemophilia A or B who have switched from prior factor or bypassing agent prophylaxis [abstract]. 2023. Publisher Full TextGoogle Scholar
- Shapiro AD, Angchaisuksiri P, Astermark J. Long-term efficacy and safety of subcutaneous concizumab prophylaxis in hemophilia A and hemophilia A/B with inhibitors. Blood Adv. 2022; 6(11):3422-3432. Google Scholar
- Mahlangu J, Lamas JL, Morales JC. Long-term safety and efficacy of the anti-tissue factor pathway inhibitor marstacimab in participants with severe haemophilia: phase II study results. Br J Haematol. 2023; 200(2):240-248. Google Scholar
- Mancuso ME, Callaghan MU, Kruse-Jarres R. Emicizumab prophylaxis in adolescent/adult patients with hemophilia A previously receiving episodic or prophylactic bypassing agent treatment: updated analyses from the HAVEN 1 study. Blood. 2017; 130(Suppl 1):1071. Google Scholar
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.