Abstract
Background
Emicizumab, a bispecific factor VIII mimetic antibody, was approved in 2018 for bleeding prophylaxis in congenital hemophilia A with or without inhibitors. Since then, several case reports and case series have described the off-label use of emicizumab in acquired hemophilia A (AHA), and data from two clinical trials were recently published (AGEHA, GTH-AHA-EMI).
Objectives
To describe the reported data on the outcomes of emicizumab, highlighting its benefit/risk profile in treatment.
Methods
We conducted a literature search in PubMed, Scopus, Cochrane, and Google Scholar up to August 2024, including all scientific articles reporting clinical outcomes of emicizumab use in patients with AHA.
Results
Thirty-two studies were included in the final review, covering a total of 171 AHA patients. The majority started emicizumab for active bleeding management and prophylaxis with various regimens. Follow-up duration and remission criteria varied. Two clinical trials supported the use of emicizumab for bleeding prophylaxis with a new dosing regimen and completion criteria. Bleeding was well managed in all cases, with no major recurrent bleeds. Some adverse events were reported : 3 cases of deep venous thrombosis, 2 cases of stroke, and 2 cases of anti-emicizumab drug antibodies developing in patients with thromboembolic risk factors.
Conclusions
Based on published data, emicizumab appears to be effective in bleeding management and prophylaxis in AHA patients, with a favorable benefit/risk profile.
Keywords: acquired hemophilia, emicizumab, review, bleeding, thrombosis
Introduction
Acquired hemophilia A (AHA) is a rare, life-threatening autoimmune bleeding disorder caused by the development of neutralizing autoantibodies against endogenous clotting factor VIII, 1 with an overall incidence of 1.5 cases per million per year.2,3 AHA primarily affects older individuals, often with cardiovascular and thromboembolism comorbidities, leading to a serious condition with a high mortality rate ranging from 8% to 48%.3,4 Additionally, several risk factors including advanced age, malignancy, FVIII levels below 1%, severe bleeding, and higher corticosteroid doses, significantly increase the incidence of early death.5,6 Younger women may also be affected during pregnancy or postpartum.2,7 Many underlying etiologies have been reported, including autoimmune disorders, malignancies, infections, and drugs. However, AHA remains idiopathic in about half of the cases, with no identifiable causes or concomitant factors. 8
Management of AHA presents a challenge to achieve bleeding control and inhibitor eradication. Bypassing agents (BPAs) such as activated prothrombin complex concentrate (aPCC) and recombinant activated factor VIIa (rFVIIa) are the first-line conventional hemostatic treatments,9–13 with a high efficacy rate of 93%. 9 Recombinant porcine factor VIII (rpFVIII) is used in for patients without cross-reactivity with an efficacity rate of 70%.9,11–13 However, these therapies are limited by the need for intravenous infusions, the lengthy period of time required to achieve remission, the risk of thrombotic complications ranging from 0% to 4.8%,2,10 and the potential development of anti-porcine antibodies when using rpFVIII. 10
According to AHA guidelines, inhibitor eradication is based on immunosuppressive therapy (IST), with corticosteroids as first-line treatment, either alone or in combination with cyclophosphamide. 13 However, achieving remission with IST takes longer and is associated with a high rate of severe side effects, particularly infections, which contribute to an overall mortality rate of up to 16%.6,14 Given the complexity and challenges of managing AHA, many studies have described the efficacy of emicizumab as a novel hemostatic approach since 2018. Emicizumab is a bispecific humanized recombinant monoclonal antibody that mimics factor VIII (FVIII) cofactor function.15–17 It binds activated factor IX (FIXa) and factor X (FX) to restore the tenase function mediated by activated FVIII (FVIIIa). 18 Emicizumab has been widely approved for bleeding prophylaxis in congenital hemophilia A (CHA) with or without inhibitors.19–21
Due to its mechanism of action, emicizumab has been used off-label as a hemostatic agent in the management of AHA patients in many parts of the world, including Europe and the United States. It was recently approved for AHA in Japan. Several recent studies, mainly case reports and case series, have described the safety and efficacy of emicizumab in AHA patients, along with available results from a clinical trial in Japan (AGEHA) 22 and the final analysis of the German-Austrian GTH-AHA-EMI trial.23,24 In this context, several considerations require special attention: indications, dosing regimens, follow-up periods, recognition of remission, interference with lab assays, concomitant use of other BPAs, thrombogenicity, adverse events, and particular situations such as surgery and childbirth. In this systematic review, we summarize the literature findings on the topic, focusing on the clinical presentations and outcomes of emicizumab, while highlighting its benefit/risk profile, limitations, and challenges in the management of AHA.
Materials and Methods
Guidelines
We conducted a systematic review on the efficacy of emicizumab as a hemostatic agent in the management of bleeding disorders related to AHA, focusing on clinical settings and outcomes based on all available evidence. The review was performed in accordance with the Preferred Reporting Items for Systematic Reviews (PRISMA) guidelines. The PRISMA checklist is provided in Annex 1.
Search Strategy
The PubMed, Scopus, Cochrane, and Google Scholar databases were searched for publications, with English language as a restriction. The search was conducted using the keywords “acquired haemophilia” OR “acquired haemophilic” OR “acquired hemophilia” OR “acquired haemophiliacs” OR “acquired factor 8 deficiency” linked to “emicizumab” OR “hemlibra,” using Boolean operators AND. The search included studies published up until 12 August 2024.
Eligibility Criteria
Two authors (IG, HB) independently conducted the search and assessed the eligibility of each article. The eligibility criteria were based on the Population, Intervention, Comparator, Outcome, and Study design (PICOS) framework (Table 1).
Table 1.
Eligibility criteria.
| Inclusion criteria | Exclusion criteria | ||
|---|---|---|---|
| P | Participate | People with acquired hemophilia A | Healthy individuals, patients with other acquired or congenital bleeding diseases |
| I | Intervention | emicizumab | Physiopathology, immunology |
| C | Comparison | - | - |
| O | Outcome | efficacy of emicizumab | - |
| S | Study | Case report, case series, conference abstract, retrospective study containing patient population >1, clinical trials | Studies without a clinical outcome, review papers, editorials |
Quality Assessment
The quality assessment of the studies was performed independently using the Joanna Briggs Institute (JBI) tool for assessing the methodological quality of case reports and case series, and the ROBINS-I tool for non-randomized studies (Annex 2)
Data Extraction
The following data were extracted from all included studies: publication information, patient age and sex, underlying disease, comorbidities, bleeding site, factor VIII levels and inhibitor titer, hemostatic and IST indication, emicizumab regimen (dose, frequency, duration), clinical response, FVIII levels upon emicizumab discontinuation, treatment downtime, time to remission, and adverse effects.
Statistical Analysis
Data are expressed as frequencies, medians, and ranges, as appropriate.
Results
A total of 508 records were identified. After removing duplicates and non-conforming papers, 32 studies including 25 full-text articles and 7 conference abstracts identified through reference checking were assessed for eligibility. The PRISMA flow chart is shown in Figure 1.
Figure 1.
Flow chart of study selection.
Study Characteristics
Given the rarity of the disease, most of the eligible studies were off-label studies conducted between 2019 and 2024 (Table 2). The use of emicizumab in AHA has been reported in 15 case reports, 7 case series from Vienna, 7 abstracts, and 2 clinical trials exploring bleed prevention in AHA. The Japanese AGEHA study combined emicizumab with IST in Cohort 1 (n = 11) and provided data on IST-ineligible patients in Cohort 2 (n = 2). The German-Austrian GTH-AHA-EMI study (N = 47) delayed IST during the initial 12-week efficacy period.
Table 2.
Review of Publications Evaluating the Emicizumab Outcomes in Acquired Hemophilia A with Patients Characteristics.
| Publication References type |
Age/gender Underlying disease |
comorbidity | Bleeding site | FVIII level Inhibitor title (BU/ml) |
Prior hemostatic agent/IST | Emicizumab indications | Dose/frequency | Efficacy | Emicizumab duration therapy | FVIII levels/inhibitor title on Emicizumab disruption | Complete remission/ periode time | Adverse effect |
|---|---|---|---|---|---|---|---|---|---|---|---|---|
|
Dane
25
Case report |
72/M AID |
CAD | hematoma | Undetectable 409 |
aPCC; rp-FVIII/ Corticosteroids, rituximab, cyclosporine, cyclophos-phamide, azathioprine, bortezomib, mycophenolate, cladribine, tacrolimus | Prophylaxis to planned PCI Allow use of DAPT following DES placement for ACS |
Standard Loading And maintenance doses |
DAT tolorated without bleeding accident | >5months | NM | NM | None |
|
Escobar
26
Case report |
90/M/idiopathic | CAD, AF, AHT, CKD | NM | Undetectable 150.4 |
NM/ Rituximab, mycophenolate | Recurrent bleeding | Modified protocole : loading dose of 1.5 mg/kg/week for two doses, then maintenance dose of 1.5 mg/kg every 21 days, then increased to 1.5 mg/kg every 14 days | Breakthrough bleed resolved with increased dose to 1.5 mg/kg every 14 days | 9 months | 10%/NM | NM | NM |
|
Escobar
27
Case report |
57/F/idiopathic | NM | NM | NM 6.67 |
NM | Recurrent bleeding | Standard Loading and maintenance doses | immediate cessation of bleeding episodes | NM | NM | NM | NM |
|
Publication
References type |
Age/gender Underlying disease |
comorbidity | Bleeding site | FVIII level Inhibitor title (BU/ml) |
Prior hemostatic agent/IST | Emicizumab indications | Dose/frequency | Efficacy | Emicizumab duration therapy |
FVIII levels/inhibitor title on Emicizumab disruption | Complete remission/ periode time | Adverse effect |
|
Knoebel
28
Case series (n = 12) |
61/M, idiopathic | IS Infection CAD LD |
haematoma, GI, post surgical site bleed | <1% 69 |
aPCC rFVIIIa rFVII/prednisone, rituximab |
Active refractory bleeding | Standard loading and maintenance doses; median number of doses 5 (range, 3-9) | No recurrent bleeding 5 days after the first dose | Median 31 days [15–79] | FVIII:h > 30%/NM | FVIII:h > 50%/NM | Died 45 days after last dose of emicizumab from peritonitis |
| 51/M idiopathic |
adiposity | Cutaneous bleed | <1% 79.4 |
None/prednisone, rituximab | Active bleeding | No recurrent bleeding | Median 31 days [15–79] | FVIII:h > 30%, NM | FVIII:h > 50%/104 days | None | ||
| 62/F, idiopathic | none | Cutaneous bleed, hematoma | 3% 3.5 |
rFVIIa, rFVIII, rp-FVIII/ prednisone, rituximab | Active refractory bleeding | No further bleeding 3 days after first dose | Median 31 days [15–79] | FVIII:h > 30%, NM | (FVIII:b > 50%/403 days | None | ||
| 79/F, idiopathic | Infection Adiposity Peripheral arterial disease |
Cutaneous bleed, haematoma, post-surgical site bleed | <1% 80.4 |
aPCC, rFVIIa /prednisone, rituximab | Active refractory bleeding | Bleeding controlled 15 days after first dose | Median 31 days [15–79] | FVIII:h > 30%, NM | (FVIII:b > 50%)/265 days | Minor stroke on day 16 after third dose of emicizumab, associted with rFVIIa used for surgery | ||
| 87/M, idiopathic | Diabetis Adiposity malignancy |
Cutaneous bleed, haematoma | <1% 17.1 |
rFVIII, aPCC/rituximab | Prophylaxis NSTEMI on aPCC |
bleeding controlled 4 days after first dose | Median 31 days [15–79] | FVIII:h > 30%, NM | (FVIII:b > 50%)/180 days | None | ||
| 82/F, idiopathic | Diabetes Adiposity malignancy |
Cutaneous bleed, haematoma, post-surgical site bleed | <1% 14.5 |
rFVIIa/rituximab | Active refractory bleeding | Bleeding controlled 2 days after first dose | Median 31 days [15–79] | FVIII:h > 30%, NM | (FVIII:b > 50%)/120 days | none | ||
| 86/F, idiopathic | Arterial fibrillation | Cutaneous bleed, haematoma, post-surgical site bleed | <1% 228.5 |
rFVIIa/prednisone, rituximab | Active refractory bleeding | Bleeding controlled 4 days after first dose | Median 31 days [15–79] | FVIII:h > 30%, NM | (FVIII:b > 50%)/110 days | None | ||
| 79/M, idiopathic | Adiposity Atrial fibrillation Peripheral arterial disease CHD |
Cutaneous bleed, haematoma, post-surgical site bleed | 5% 3.6 |
rFVIIa, rFVIII/ Prednisone, rituximab | Active refractory bleeding | Bleeding controlled 3 days after first dose | Median 31 days [15–79] | FVIII:h > 30%, NM | (FVIII:b > 50%)/18 days | None | ||
| 67/F, idiopathic | depression | Cutaneous bleed, haematoma | <1% 27.6 |
rFVIIa/ Prednisone, rituximab | Active refractory bleeding | Bleeding controlled 3 days after first dose | Median 31 days [15–79] | FVIII:h > 30%, NM | (FVIII:b > 50%)/187 days | None | ||
| 65/M, idiopathic | Diabetes Adiposity LD |
Cutaneous bleed, haematoma | 11% 8.9 |
rFVIII/rituximab | Active refractory bleeding | NM | Median 31 days [15–79] | FVIII:h > 30%, NM | (FVIII:b > 50%)/54 days | None | ||
| 69/M, idiopathic | AF LD |
Mucocutaneous bleed, haematoma, haematuria, joint bleed, post-surgical site bleed | <1% 2000 |
rFVIIa/ Prednisone, rituximab | Active refractory bleeding | Bleeding controlled 3 days after first dose | Median 31 days [15–79] | FVIII:h > 30%, NM | (FVIII:b > 50%)/>175 days | None | ||
| 79/F, idiopathic | Malignancy depression | Cutaneous bleed, haematoma | 1% 8.8 |
rFVIIa/ Prednisone, rituximab | Active refractory bleeding | Bleeding controlled 3 days after first dose | Median 31 days [15–79] | FVIII:h > 30, NM | (FVIII:b > 50%)/27 days | None | ||
|
Jena
29
Case report |
78/M/cancer | AHT | Cutaneous bleed, GI bleed | 2.5% 34 |
rFVIII, aPCC/ Steroids, cyclophosphamide | Active refractory bleeding | NM | No further bleeding | NM | NM | NM | NM |
|
Chen
30
Case series (n = 8) |
85/F, idiopathic | NM | Cutaneous bleed, haematoma | 3% 40 |
rFVIIa/ rituximab | Active refractory bleeding | standard loading and maintenance dose of 3 mg/kg /2 weeks | No further bleeding | 3 months | NM | NM | None |
| 78/M AID |
NM | Cutaneous bleed, haematoma | <1% 33 |
None/rituximab, prednisone | Active refractory bleeding | standard loading dose | No further bleeding | 4 weeks | 29%/NM (71 days after starting emicizumab) | NM | None | |
| 85/F, idiopathic | NM | Cutaneous bleed, haematoma | 2% 9.5 |
rFVIIa/ rituximab, prednisone | Active refractory bleeding | standard loading dose | No further bleeding | 4 weeks | 86%/NM (91 days after starting emicizumab) | NM | None | |
| 69/F, idiopathic | NM | Post-surgical site bleed | <1% 107.5 |
None/ rituximab | Active refractory bleeding | standard loading dose | No further bleeding | 4 weeks | NM | NM | None | |
| 87/M, idiopathic | NM | hematuria | 1% 9.5 |
rFVIIa/ rituximab | Active refractory bleeding | standard loading dose | recurrent haematuria required embolisation | 4 weeks | NM | NM | None | |
|
Publication
References type |
Age/gender
Underlying disease |
comorbidity | Bleeding site |
FVIII level
Inhibitor title (BU/ml) |
Prior hemostatic agent/IST | Emicizumab indications | Dose/frequency | Efficacy | Emicizumab duration therapy | FVIII levels/inhibitor title on Emicizumab disruption | Complete remission/ periode time | Adverse effect |
| 93/F, idiopathic | NM | Cutaneous bleed | 1% 9.2 |
none/ rituximab, prednisone | Active refractory bleeding | standard loading dose | No further bleeding | 4 weeks | NM | NM | None | |
| 54/M, malignancy | NM | Haematoma, bronchial artery bleed | 2% 10 |
rFVIIa/ rituximab | Active refractory bleeding | standard loading dose | No further bleeding | 4 weeks | NM | NM | None | |
| 47/F, malignancy | NM | Cutaneous bleed, epistaxis | 9% 26 |
none/ rituximab, prednisone | Active refractory bleeding | standard loading dose | No further bleeding | 4 weeks | NM | NM | None | |
|
El banna
31
Case report |
87/F/idiopathic | AF | GI bleed, chest and pelvic hematomas | <1% >100 |
aPCC 50 units/kg every 12 h for 2 weeks./ none | Bleeding prophylaxis | Standard loading and maintenance dose | No further bleeding | >2 months | NM | NM | None |
|
Ganslmeier
32
Case report |
59/M/idiopathic | COPD, T2DM, obesity, AHT | retroperitoneal haemorrhage | Undetectable 136 |
rFVIIa, ac tranexamique/prednisone, cyclophosphamide | Recurrent bleeding | Loading dose unspecified Maintenance dose 2.7 mg/kg /4weeks |
No further bleeding/7 days | >10 months | NM | NM | Eczema in the neck with the second injection |
|
Hess
33
Case report |
91/M/idiopathic | AHT, benign prostatic hyperplasia, AF, mitral valve replacement | Hematuria, psoas hematomas | <1% 44 |
rFVII 90 μg/kg/prednisone, cyclophosphamide | Recurrent bleeding | Standard loading then maintenance dose of 1.5 mg/kg every 2 weeks. | No further bleeding | 6 months | 86/1.9 | NM | none |
|
Publication
References type |
Age/gender
Underlying disease |
comorbidity | Bleeding site |
FVIII level
Inhibitor title (BU/ml) |
Prior hemostatic agent/IST | Emicizumab indications | Dose/frequency | Efficacy | Emicizumab duration therapy | FVIII levels/inhibitor title on Emicizumab disruption | Complete remission/ periode time | Adverse effect |
|
Mohnel
34
Case report |
83/M/idiopathic | congestive heart failure, AF, CKD, DVT, PE | GI bleed, haematoma, mucocutaneous bleed | <1% 97 |
rFVIII, aPCC, FXIII, and fibrinogen/ prednisolone ; rituximab ; IVIG | Recurrent bleeding | modified dose at 3 mg/kg for first dose, 1.5 mg/kg for second dose 1 week later and 1.5 mg/kg for third dose at 20 days post-first dose | No further bleeding | 20 days | 10%/ 2 (35 days after last dose) | NM | Died 36 days after last dose of emicizumab due to an arrhythmic event |
|
Flommersfeld
35
Case report |
21/F AID |
NM | post chirurgical bleed, hematoma | <0.25% 40 |
rFVII/ Dexamethasone ; cyclophosphamide, ofatumumab, bortezomib and daratumumab ; ITT with IVIG and high-dose FVIII substitution | Recurrent bleeding | Standard loading and maintenace doses | led after tooth extraction requiring rFVIIa for 7 days | <3months | NM | NM | NM |
|
Chen
36
Case series (n = 4) |
57/F, idiopathic | T2DM Hyperlipidaemia, obsesity, hypothyroidism |
hematoma | <1% 10 |
rpFVIII ; rFVIIa/ Rituximab ; cyclophosphamide | Recurrent bleed | Standard loading and maintenance doses | Traumatic elbow bleed resolved with hemostatic concentrate factor | 61 days | normolized FVIII and eradication of inhibitor | CR/NM | none |
| 67/M, idiopathic | T2DM, CAD, AHT, gout, MGUS, dementia | Haematoma, GI bleed, epistaxis | <1% 162 |
rpFVIII/ rituximab | Recurrent bleed | Standard loading and maintenance doses | No reccurent bleed | 216 days | normolized FVIII and eradication of inhibitor | CR /NM | none | |
| 74/M, idiopathic | T2DM, COPD, diverticulitis | Haemarthrosis, haematoma | 1% 85 |
rpFVIII/ rituximab | Recurrent bleed | Standard loading and maintenance doses | No reccurent bleed | 102 days | normolized FVIII and eradication of inhibitor | CR/NM | none | |
| 68/M, idiopathi | CAS, AHT | Hematoma | <1% 9 |
rpFVIII/prednisone Rituximab ; cyclophosphamide | Recurrent bleed | Standard loading and maintenance doses | No reccurent bleed | >211days | Still on emicizumab | Still on emicizumab | none | |
|
Chen
37
Case series (n = 11) |
84/F idiopathic |
NM | Ecchymosis, hematoma | 3% 40 |
None/rituximab | Active refractory bleeding | 3 mg/kg weekly for 4 weeks then3 mg/kg every 2 weeks to complete 3 months | No reccurent bleed | 4 month | FVIII activity to > 50% with a negative inhibitor | CR/NM | none |
| 77/M idiopathic |
Rheumatoid arthritis, AF | Traumatic hematoma hematuria | <1% 33 |
rFVIIa/rituximab, prednisone | Active refractory bleeding | Standard loading dose | No reccurent bleed | 1 month | FVIII activity to > 50% with a negative inhibitor | CR/NM | none | |
| 84/F idiopathic |
Factor V Leiden | ecchymoses, abdominal hematoma requiring artery embolization |
2% 9.5 |
rFVIIa/rituximab, prednisone, | Active refractory bleeding | Standard loading dose | No reccurent bleed | 1 month | FVIII activity to > 50% with a negative inhibitor test | CR/NM | none | |
| 68/F malignancy |
Recurrent liposarcoma | post-procedual bleeding | <1% 107.5 |
rFVIIa/rituximab | Active refractory bleeding | Standard loading dose | No reccurent bleed | 1 month | FVIII activity to > 50% with a negative inhibitor | CR/NM | none | |
| 87/M idiopathic |
Benign prostatic hyperplasia | spontaneous hematuria requiring prostate artery embolization. | 1% 9.5 |
rFVIIa/rituximab | Active refractory bleeding | Standard loading dose | No reccurent bleed | 1 month | FVIII activity > 50% but with detectable inhibitor | PR /NM | none | |
| 93/F idiopathic |
AF | traumatic ecchymoses | <1% 9.2 |
None/rituximab | Active refractory bleeding | Standard loading dose | No reccurent bleed | 1 month | FVIII activity to > 50% with a negative inhibitor | CR/NM | none | |
| 54/M malignancy |
Tonsillar squamous cell carcinoma | hematoma hemoptysis. | 2% 10 |
rFVIIa/ritixumab, | Active refractory bleeding | Standard loading dose | No reccurent bleed | 1 month | FVIII activity > 50% but with detectable inhibitor | PR/NM | none | |
| 47/F malignancy |
acute myeloid leukemia | hematoma ecchymoses requiring vascular embolization | 9% 26 |
None/rituximab, prednisone | Active refractory bleeding | Standard loading dose | No reccurent bleed | 2 weeks (insurrance approval) | FVIII activity to > 50% with a negative inhibitor | CR/NM | none | |
| 70/F idiopathic |
venous thromboembolism (FVL+) | Post-surgery hematoma requiring artery embolization | 6% 47 |
Acide tranexamique /rituximab | Active refractory bleeding | Standard loading dose | No reccurent bleed | 1 month | FVIII activity to > 50% with a negative inhibitor | CR/NM | none | |
| 72/M idiopathic |
NM | hematuria hemarthrosis | <1% 103 |
rFVIIa/rituximab | Active refractory bleeding | Standard loading dose | Rebleeding occurred on day 24 after initiating emicizumab | 1 month | NM | Refractory disease | none | |
| 81/M Covid-19 |
COVID-19 vaccination | Traumatic ecchymoses | <1% 84 |
None/rituximab | Active refractory bleeding | Standard loading dose | No reccurent bleed | 1 month | FVIII activity to > 50% with a negative inhibitor test in the absence of haemostatic and IST | CR/NM | none | |
|
Publication
References type |
Age/gender
Underlying disease |
comorbidity | Bleeding site |
FVIII level
Inhibitor title (BU/ml) |
Prior hemostatic agent/IST | Emicizumab indications | Dose/frequency | Efficacy | Emicizumab duration therapy | FVIII levels/inhibitor title on Emicizumab disruption | Complete remission/ periode time | Adverse effect |
|
Poston
38
Case series (n = 24) |
Median 73 [34–87] M > F AID Cancer Peripartum |
Metabolic syndrome Vascular disease Prior venous thrombosis |
Hematuria Hemarthrosis Retroperitoneal GI |
<1% (1%-28%) 54 (0.8–749) |
rFVII, FEIBA, recombinant porcine/glycocorticoids (86%), cyclophosphamide (17%), mycophenolate mofetil (13%), daratumumab (4%). | Bleeding prophylaxis | Standard loading and maintenance doses | Bleeding resolved Recurrent bleeding (hematuria, GI) |
13 days (1–217) | NM | NM | DVT (1 case) 2 Death not attribuated to emicizumab |
|
Happaerts
39
Case report |
75/M MAI, Covid-19 vaccine (astrazeneca) |
CKD, AHT, hypertension, complicated IDDM | multiple hematomas, hemorrhagic bullous pemphigoid, and GI ulcer | Undetectable 135 | rFVIIa/ rituximab, methylprednisolone | Active bleeding, started concurrent with rFVII | 3 mg/kg, 2gifts | No further bleeding events | 15 days | 13%/75 | NM | death unrelated to emicizumab |
|
Karthick
40
Case report |
84/M Covid |
AHT | Extensive ecchymotic patches | <1% 1.8 |
FFP, rFVIIa, FEIBA/Azathioprine, Rituximab | Active refractory bleeding | One dose (210 mg) | No further bleeding | 1 day | NM | NM | Death related to cardiac arrest after covid treatement |
|
Publication
References type |
Age/gender
Underlying disease |
comorbidity | Bleeding site |
FVIII level
Inhibitor title (BU/ml) |
Prior hemostatic agent/IST | Emicizumab indications | Dose/frequency | Efficacy | Emicizumab duration therapy | FVIII levels/inhibitor title on Emicizumab disruption | Complete remission/ periode time | Adverse effect |
|
Nishiki
41
2 cases report |
82/F/NM 59/F/NM |
Diabetes NM |
NM NM |
<1% 58 <1% 33 |
rFVIIa aPCC |
Patient ineligible for IST Patient ineligible for IST |
6 mg/kg on Day 1 and 3 mg/kg on Day 2 followed by 1.5 mg/kg weekly from Day 8 6 mg/kg on Day 1 and 3 mg/kg on Day 2 followed by 1.5 mg/kg weekly from Day 8 |
No further bleeding No further bleeding |
7.1 weeks 16.3 weeks |
NM NM |
FVIII > 50% on day 64 FVIII > 50% on day 116 |
Rash Rash |
|
Hansenne
42
2 cases report |
73/M Idiopathic 93/M Cancer |
gout, AHT, chronic alcohol consumption prostatic adenocarcinoma, AHT |
Hematomas hematomas |
<1% 15.2 1% 11 |
rFVIIa / methylprednisolone, rituximab, cyclosporine none/methylprednisolone, rituximab |
Prophylaxis Active bleeding |
Standard loading dose then 6 mg/kg once Standard loading and maintenance doses |
No recurrent bleeding No recurrent bleeding |
NM NM |
NM 63% |
NM NM |
None none |
|
Hayden
43
Case report |
90/M idiopathic |
NM | hematoma | <1% 3315 |
rFVIIa/prednisone, ritixumab | Active refractory bleeding | Standard loading dose then at 1.5 mg/kg every 3 weeks | No recurrent bleeding | NM | 29% | NM | none |
|
Gelbenegger
44
Case report |
75/M Covid-19 |
myocardial infarction with coronary stenting, AHT, hyperlipidaemia, infrarenal aortic aneurysm | hematoma | <1% 41.8 |
rFVII/prednisone | Bleeding prophylaxis | 3 mg/kg weekly for the first 3 weeks, thereafter with 1.5 mg/kg every 2–4 weeks | No further bleeding 4 days after the first injection | 105 days | FVIII : h : 10–30% | CR : FVIII : b > 50% negative inhibitor, no active bleeding after stopping any haemostatic drug for >24 h | none |
|
Kaunchale
45
Case report |
60/M/NM | T2DM, asthma, | Extensive bruising | 1.3% 8 |
FEIBA/steroid | Bleeding prophylaxis | 1.5 mg/Kg (one dose) | No further bleeding | 1 day | NM | NM | none |
|
Yates
46
Case report |
83/M /NM |
AF, DIT Infection AHT, LGLPD |
Spontaneous ecchmosis, Abdominal hematoma |
<1% 197 |
rFVIIa, tranexamic acid, aPCC, /prednisone, cyclophosphamide, rituximab | Bleeding prophylaxis | Loading dose 1.5 mg/kg | Recurrent bleeding 5 days after emicizumab initiation needed rFVIIa | 224 days | 34% 0.6 |
NM | none |
|
Shima et al
23
Clinical Trial (n = 11) |
6 M/6F /median age 76 (50–92) |
AID malignancies. | NM | 1 (<0.8–36.6) 40.5 (1–149) | none/ Prednisolone in all with cyclophosphamidein 3and cyclosporine in one | Bleeding prophylaxis | 6 mg/kg (day 1), 3 mg/kg (day 2) + 1.5 mg/kg/weak (from day 8) | No further bleeding within 1 week after the first loading dose 5 minor bleeds in 2 patients | 44.5 (8–208) | FVIII >50% and no bleeds within 72 h | Asymptomatic DVT related to emicizumab ADA |
|
| Knoebl et al 47 Case serie (n = 20) | 11 M/9F 79[ 51–87] |
NM | NM | <1% 69 (3–2300) |
rFVIIa/corticosteroids, rituximab | Active refractory bleeding | Standard loading dose then 1.5 mg/kg/2–4 wks | No further bleeding | 115 days | FVIII > 50% | FVIII > 50% | none |
|
Publication
References type |
Age/gender
Underlying disease |
comorbidity | Bleeding site |
FVIII level
Inhibitor title (BU/ml) |
Prior hemostatic agent/IST | Emicizumab indications | Dose/frequency | Efficacy | Emicizumab duration therapy | FVIII levels/inhibitor title on Emicizumab disruption | Complete remission/ periode time | Adverse effect |
|
Latef et al
48
Case report |
Middle age/F/infection | HIV | NM | NM | BPA /Corticosteroids, cyclophosphamide | Recurrent bleeding | Standard loading and maintenance doses | No further bleeding | NM | NM | NM | none |
|
Crossette et al
49
Case report |
F/post-partum | SARS-Covid/vaccination | NM | NM | rpFVIII, BPA/Corticosteroids, azathioprine | Recurrent bleeding | Standard loading and maintenance doses | No further bleeding | NM | NM | NM | none |
|
Al banaaet al
50
Case report |
M/79/idiopathic | AF, T2DM RA |
Musculoskeletal bleeds, ecchymoses, hematoma | <1% 627 |
FVIIa, rpFVIII/prednisone | Recurrent bleeding | Standard loading and maintenance doses then 1.5 mg/kg/2wk (reduced after DVT and restrat apixaban) | No further bleeding | 9 months | NM | NM | DVT |
|
Tiede et al
24
Clinical Trial (n = 47) |
23F/24M 76 years [66–80] Autoimmune diseases malignancies postpartum |
D2T Cardiac disorders Renal disordres |
NM | 1.4 [0.3–5.6] 12.2 [4.1–47.2] | NM/None for the first 11 weeks then 29 received IST | Bleeding prophylaxis | 6 and 3 mg/kg on day 1 and 2, 1.5 mg/kg weekly until week 12 | No further bleeding in 33 patients 7 patients had one bleed, 6 had 2 bleeds and 1 had 3 bleeds |
12 weeks and Follow up until 24 weeks | NM | NM | One Stoke 4 death not related to emiciczumab |
|
Engelen et al
51
Case serie (n = 7) |
NM | NM | hematoma intramuscular bleeding haematuria gastro-intestinal bleeding | 0%[0–1] 182 BU/mL[ 104–228] |
Three patients received activated FVIIa/IS | Bleeding prophylaxis | 3 mg/kg for 4 weeks then every 2 weeks until inhibitor regression | No further bleeding | NM | NM | NM | none |
|
Ahmed et al
52
2 cases reports |
M/66/idiopathic | Gout, hypertension | Extensive ecchymosis hematoma | <1% 430 BU/mL |
Factor VIIa, pFVIII/prednisone,retixumab,cyclophosphamide | Bleeding prophylaxis | loading dose 3 mg/kg/2–3weekly maintenance doses 1.5 mg/kg/3weeks |
No further bleeding | 3 months | FVIII = 35% Inhibiteur = 0 |
1 year | none |
| M/64/idiopathic | T2DM/ CKD/ gout | Ecchymosis retroperitonealhematoma |
<1% 353BU/ml |
Factor VIIa/ prednisone,retixumab,cyclophosphamide | Bleeding prophylaxis | loading dose 3 mg/kg/2–3weekly maintenance1.5 mg/kg/3weeks |
No further bleeding | 3 months | FVIII > 50% Inhibiteur = 0 |
1 year | none | |
|
Shima et al
22
Clinical trials (2 cohorts) Cohort 1 23 (n = 11) IST ligible patients Cohort2 (n = 2) IST-ineligible patients |
Cohort 2 F/82 |
T2D | Major bleeds | <1% 33 |
None/none | Delayed IST | 6 mg/kg on Day 1, 3 mg/kg on Day 2, and 1.5 mg/kg once weekly from Day 8 | 2 major bleeds stopped within the following day 3 | 64 days | M | NM | |
| F/59 | Hashimoto's disease multiple myeloma | Major bleeds | <1% 58 |
aPCC/none | IST not tolorated | 6 mg/kg on Day 1, 3 mg/kg on Day 2, and 1.5 mg/kg once weekly from Day 8 | Bleeding stopped on day 15 | 450 days | NM | NM | ADA | |
| Cohort 1 F/88 |
NM | Major bleeds | NM | rFVIIa/low dose of IST | Reduced IST dose to avaoid side effect | 6 mg/kg on Day 1, 3 mg/kg on Day 2, and 1.5 mg/kg once weekly from Day 8 | Bleeds decreased in the latter half of emicizumab periode of treatment | 639 days | NM | NM | ADA |
AID: auto-immune disease; ACS: acute coronary syndrom;ADA: ant-emicizumab drug antibodies, aPCC: activated prothrombin complex concentrate; AHT: arterial hypertension; AF: arterial fibrillation; BPA: bypassing agents, CAS: carotid artery stenosis; CAD: coronary artery disease ; CKD : chronic kidney disease; COPD: chronic obstructive pulmonary disease ; CHD : chronic heart disease ; CR : complete remission ; DES : drug eluting stent ; DAPT : dual antiplatelet therapy ; DIT : drug induced thrombocytopenia ; DVT :deep vein thrombosis ; F : female ; FVIII:h : factor VIII level using human reagent ; FFP :fresh frozen plasma ; FVL: V leiden factor ; GI = gastrointestinal ; IDDM : insulin-dependent diabetes mellitus ; IST : immunosupression therapy ; IS : immunosupression ; IBD : inflammatory bowl disease ; ITT : induce tolerated therapy ; IVIG :intravenous immunoglobulin ; LGLPD : low grade lymphoproliferative disorder ; LD : lung disease ;M = male ; MGUS : monoclonal gammapathy of underterminated significance ; NSTEMI : non-ST elevation myocardial infarction ; NM : not mentionned ; PCI : percutaneous coronary intervention ; PE : pulmonary embolism ; PR : partial remission ; rpFVIII :recombinant porcine FVIII ; rFVIIa : recombinant activated factor FVII ; T2DM :type 2 diabetes mellitus ; Standard loading dose : 3 mg/kg subcutaneous weekly for 4 doses ; Maintenance dose : 1.5 mg/kg weekly;PR : FVIII > 50% with no active bleeds, CR : PR with inhibitor eradication.
Population Characteristics
One hundred seventy-one AHA patients were treated with emicizumab. The median age was 68 years (range: 21-93), with 55.8% being male. AHA was considered secondary to malignancy, autoimmune disease, COVID-19, and HIV infection in 13.4%, 7.7%, 7.6%, and 2% of patients, respectively.23–25,29,30,35,37–40,42,44,48,49,52 It was idiopathic in 69.8% of patients.25–28,30–34,36,37,42,43,50,52 Every patient had at least one comorbidity with a higher frequency of metabolic syndrome conditions and commonly arterial fibrillation and coronary artery disease.
Clinical Bleeding Symptoms
AHA patients presented with a range of clinical signs of hemorrhagic diathesis. The most common symptoms were mucocutaneous bleeds (74.1%), hematomas (72.2%), post-surgical bleeds (18.5%), and less frequently hemarthrosis (5.5%).23,25,28–31,34–38,42–44,46,50–52 The symptoms were spontaneous and occurred at multiple sites. Additionally, most of the patients met the criteria for severe bleeding, and four patients required an embolization procedure. 37
Laboratory Methods
In 68.5% of patients, FVIII levels were undetectable or lower than 1% at the onset of AHA diagnosis or emicizumab initiation.7,22,23,25,26,28,30–34,36–40,42–44,46,50–52 FVIII levels were measured using a chromogenic assay with either bovine coagulation factors23,32–37,39,42–44,47,50,52 or human reagents28,42,44,47,51 Shima et al described a new laboratory tool based on a one-stage clotting assay with emicizumab in plasma samples neutralized by adding two anti-emicizumab idiotype monoclonal antibodies ex vivo (OSAwEN).22,23 Inhibitor levels ranged between 1 and 2300 Bethesda units per milliliter (BU/ml) (Table 2). Plasma emicizumab concentration was assessed in three studies.23,26,35
Hemostatic Treatement and Antibody Eradication
Of 171 patients, 65.5% received initial hemostatic treatement prior to emicizumab initiation.22,25,28–52 Hemostatic agents were not required in 19.4% patients.22,23,26–28,30,37,41,42
The most commonly used hemostatic agent were rFVIIa (56.6%), human or porcine FVIII (23.8%) and aPCC (15.9%).24,25,28–36,38–47,49–52
Immunosuppressive therapy was used in 109 patients.23,25,28–30,32–34,36–40,42–50 Prednisolone (75%) and rituximab (75%) were the main agents used for antibody eradication followed by cyclophosphamide (14%). The combination of rituxumab and prednisolone was observed in 27.5% of the cases.
Emicizumab Indications
Emicizumab was used as a second-line therapy for active bleeding refractory to BPA (52/171, 30.4%) and for recurrent bleeding (13/171, 7.6%).26–30,32–37,40,43,47–50 Bleeding prophylaxis was the primary indication of emicizumab (n = 101/171, 59%).22–25,28,31,38,42,44–46,51,52 Some manuscripts reported the use of emicizumab in active bleeds as a firstline treatment, either alone (4.6%)28,30,37 or in combination with rFVIIa (1.7%). 39 Additionally, emicizumab was initiated in cases of IST failure or ineligibility (6%),22,23,41 to minimize immunosupression and BPA adverse effects22,24,28,38 and in case of anti-platelet therapy initiation. 25
Emicizumab Regimen (Dose, Frequency, Plasma Concentration, Duration Therapy)
The standard emicizumab loading dose conventionally used in congenital hemophilia A (3 mg/kg/week for 4 weeks) was initiated in 99 out of 171 patients25,27,28,30,31,33,35–38,42–44,47–51 while 69 patients received a modified loading dose regimen.23,24,26,34,39–41,45,46,52
Two recent clinical trials employed a new regimen based on a higher loading dose distributed over 2 days (6 mg/kg on day 1 and 3 mg/kg on day 2, followed by 1.5 mg/kg from day 2 to day 7), then weekly maintenance doses from day 8.22,24 This accelerated regimen was indicated for bleeding prophylaxis in patients with or without IST. The maintenance dose was continued in 149 patients, including 130 on the standard maintenance dose (1.5 mg/kg weekly)22–25,27,28,30,31,35–38,42,44,47–52 and 8 on a modified regimen26,32–34,41–43 The maintenance doses of 3 mg/kg every 2 weeks or 6 mg/kg every 4 weeks, derived from licensed regimens for CHA, were applied in 11 patients. Twenty-two patients did not receive any maintenance dose.30,37,39,40,42,45,46 The duration of emicizumab therapy varied widely, ranging from 1 day to over 224 days.
Different emicizumab plasma concentrations, ranging from 9 to 51 µg/ml, were reported to be effective in stopping bleeding.23,26,35 This variation depends on the dosing regimen and the timing of concentration measurement.
Follow up Duration of Emicizumab Therapy
Follow-up period was reported in 9 studies23,24,26,28,30,31,33,42,50 ranging from 14 days to over 10 months.
Emicizumab Efficacy
All cases reported the effectiveness of emicizumab in resolving active or recurrent bleeding, with a median time of 5 days (range: 0–15).23,32,37 Bleeding was controlled after the first emicizumab loading dose in 62.8% of patients, with a median time of 4.5 days (range: 1–15). However, rebleeding occurred in 40 cases, of which 7 required additional hemostatic agents.23,24,35–38,46
Remission Criteria
Eight studies reported data on remission time.22,24,28,36,37,41,44 Remission criteria were heterogeneous, based on FVIII levels using different reagents, inhibitor levels, and clinical bleeding signs. Partial remission was defined as FVIII levels >50% with no active bleeding. 37 Complete remission (CR) was defined by the following criteria: (1) no bleeding without hemostatic treatment for at least 24 h; (2) FVIII levels >50%; and (3) a negative inhibitor test.37,44 CR was achieved in a median of 96.5 days (range: 9–270).
Emicizumab Adverse Events
Adverse events were proclaimed in 10 cases : 3 cases of deep vein thrombosis (DVT),23,38,50 2 cases of stroke,24,28 one case of atopic eczema, 32 2 cases of rash 41 and 2 cases of developed anti-emicizumab drug antibodies (ADAs) without a clear pharmacokinetics impact.22,23 Thrombotic events were observed due to the concomitant use of bypassing agents, including one case of stroke and two cases of deep vein thrombosis (DVT). No case of death was related to emicizumab.
Emicizumab Disruption Time
Emicizumab disruption time varied from 1 to 99 days with a median of 58 days. FVIII levels after discontinuation of the first dose of emicizumab ranged from 10% to 123%.
Discussion
The introduction of emicizumab has significantly transformed the treatment approach for CHA in very young, untreated patients with severe diseases, as demonstrated by the recent HAVEN 7 clinical trial. 53 This advancement hints at a similar transformative potential for managing AHA. This systematic review has consistently highlighted the efficacy of “off-label” emicizumab use in AHA patients, with partial support from two clinical trials.23,24 “Although the included studies were assumed to be of rigorous methodological quality, this assessment is limited by the fact that 7 of 32 papers were only available as abstracts. This lack of full data hinders a thorough evaluation of their methodological rigor and may influence the interpretation of the results.” The effectiveness of emicizumab still has to be considered with caution given the small sample size and the absence of randomized clinical trials. However, the strict inclusion/exclusion criteria and the validated tools used to mesaure the outcomes ensure high reliability and reproductibility of data supporting the validity and the strength of the findings. In future search, it would be useful to replicate clinical trials with a large sample and with randomized series.
Emicizumab Indications
Emicizumab has shown promise as a second-line therapy for active or recurrent bleeding refractory to conventional hemostatic agents. Interestingly, bleeding was controlled from the first emicizumab loading dose in 62.8% of patients,23,28,31,32,34,37,42 with a median time of 4.5 days. Few cases reported the use of emicizumab as a first-line therapy alongside IST.28,30,37,42 Moreover, thrombin generation studies have shown that emicizumab mimics FVIII activity levels of 10 to 15 IU/dL, improving the bleeding phenotype from severe to mild.20,54 Despite this, additional therapy with BPA is still needed for breakthrough bleeding or invasive procedures. 55 Due to its pharmacokinetic and pharmacodynamic properties, emicizumab is not intended for treating acute bleeds and BPA remain preferred for their short-life time and easier monitoring 13
Emicizumab, approved for bleeding prophylaxis in CHA with and without inhibitors,19,56 may also be highly beneficial for AHA patients who have a greater risk of bleeding. AGEHA trial have shown that emicizumab improves hemostasis and significantly reduces bleeding rates with an accerelated loading dose, 23 even in patients without concomitant IST. 22 More recently, the GTH-AHA trial showed a significant reduction in bleeding rate (0.04) in patients without concomitant IST compared to the historical rate of (0.15) documented in patients with IST.24,57 These data suggest that emicizumab alone can effectively prevent most bleeding in patients resistant to or slowly responding to IST.
During the COVID-19 pandemic, when the impact of IST can be critical, emicizumab was considered the only option for bleeding prophylaxis when inhibitor eradication was not achievable.37,39,40,44,49
Emicizumab Regimen
The dosing regimen for emicizumab varied significantly due to differences in bleeding risk and patient phenotypes. The GTH-AHA working group recommends tailoring emicizumab loading regimens to specific patient need. 58 The accelerated dosing regimen used in the AGEHA and GTH-AHA-EMI trials—starting with 6 mg/kg on day 1 and 3 mg/kg on day 2—achieves steady-state levels within one week, with a trough concentration of 30 μg/mL.23,24 These studies reported promising results, including low bleeding (0%-4%) and thrombotic event rates (2%-4%) over a 12-week period.23,24 This regimen should be considered for rapid prophylaxis from the time of diagnosis. 58
Thus, higher loading doses with a reduced duration could enhance the efficacy of emicizumab, leading to an earlier clinical response due to its long half-life (approximately 30 days), even if endogenous FVIII levels remain low and inhibitor eradication is not achieved.
The saturation regimen of 3 mg/kg once a week for 4 weeks, as used in the HAVEN study for CHA patients, achieves steady-state levels of emicizumab within 4 weeks, with a trough concentration of 20 μg/ml.28,59 This lower dosing could be a viable alternative for managing AHA patients with a low bleeding tendency,52,58 even in the presence of high inhibitor levels. Observations from CHA studies suggest that even lower emicizumab plasma concentrations can be effective for bleeding prophylaxis.60,61
The recommended maintenance dose of emicizumab was 1.5 mg/kg once per week. 58 Longer dosing intervals (3 mg/kg/2weeks or 6 mg/kg/ 4weeks) may be applied for resistant or ineligible IST patients needed a long remission time. 58 Emicizumab should be discontinued once remission is ashieved. 58 Earlier discontinuation can be considered in stable patients achieving FVIII > 30%. 58 The optimal timing for discontinuation of emicizumab needs further investigation.
Emicizumab Versus IST
The risk of potentially fatal bleeding complications justifies the early initiation of IST to promptly eradicate autoantibodies. 13 Immunosupression should be administered based on the patient's physical status. 58 For ineligible patients, emicizumab may be used to delay IST initiation to avoid infection while being protected against recurrent bleeding risks.24,41,62 Hart et al compared the outcomes of GTH-AH 01/2010 6 (IST without hemostatic prophylaxis) and GTH-AHA-EMI 24 studies (hemostatic prophylaxis with emicizumab without IST) and concluded that using emicizumab instead of IST during the early phase of AHA diagnosis (within 12 weeks) reduced bleeding and fatal infections and improved overall survival. 57
Patients treated with IST experienced a higher bleeding rate compared to those treated with emicizumab during the first 5 weeks. The mean bleeding rate decreased from 0.128 to 0.042 with emicizumab, showing a significant rate ratio of 0.325 (95% CI, 0.182–0.581; P < .001). 57 Despite this, bleeding can still occur, and IST may be needed to further reduce bleeding risk by achieving partial remission (PR). Most patients received IST after week 12 without any infections by week 24. 57 This fact was supported in AGEHA results suggesting that delaying IST may reduce risks.22,57
An ongoing trial in the United States (AHA-EMI, NCT05345197) aims to evaluate the risk/benefit ratio of early IST within the first 12 weeks alongside an accelerated or modified emicizumab dosing regimen.
Survival rates at 24 weeks were significantly higher with emicizumab (90%) compared to IST (76%). 57 Emicizumab prophylaxis offers greater flexibility in timing IST, potentially improving the risk/benefit ratio of less intensive treatments.
Emicizumab and Antithrombotic Therapy
Recent data indicate that antithrombotic therapy can be safely used in AHA patients on emicizumab, as it reduces bleeding risks.24,25,46 EHA-ISTH-EAHAD-ASO guidelines allow for oral anticoagulants or antiplatelet drugs in CHA patients with clotting factor levels above 20 IU/dL, though careful monitoring is required. 63 These recommendations could be adapted for AHA patients, taking into account their higher thrombotic risk compared to CHA patients.
Emicizumab Laboratory Monitoring
The use of emicizumab requires specialized laboratory expertise due to its interference with standard clotting assays. Emicizumab bridges FIXa and FX, restoring intrinsic tenase function usually mediated by FVIIIa, which is deficient in hemophilia patients. However, this mechanism can artificially shorten the aPTT-based FVIII activity test, leading to falsely normal results at low emicizumab concentrations due to an overestimation of FVIII levels. 64
Chromogenic assays using bovine-derived reagents, are unaffected by emicizumab and are recommended for FVIII activity and inhibitor titers monitoring to achieve remission.18,58,65
Chromogenic FVIII testing with human-derived factors effectively reflects emicizumab's hemostatic activity and had a good correlation with emicizumab plasma level.18,64
FVIII inhibitors are accurately detected with modified Bethesda assays or ELISA tests, both unaffected by emicizumab. 65
The OSAwEN method provides a viable alternative for monitoring emicizumab in vivo. Shima et al found a correlation between OSAwEN method and chromogenic bovine assay which could serve as complementary tool to detect low inhibitor titers. 23 However, it's crucial to recognize that the diagnostics for AHA and the monitoring of emicizumab therapy are not routinely accessible in many healthcare institutions.
Remission Criteria
Recent clinical trials, including AGEHA and GTH-AHA-EMI, have established remission criteria and follow-up protocols for emicizumab in AHA.22,24 Remission is typically defined by the normalization of FVIII activity levels and the eradication of FVIII inhibitors. The AGEHA trial suggests that patients can be considered for remission once FVIII levels consistently exceed 30% and bleeding episodes are controlled without additional hemostatic agents. The GTH-AHA-EMI trial further emphasizes the importance of sustained FVIII activity and the absence of bleeding events as key remission indicators. Follow-up duration in these trials generally extends for 12 to 24 weeks, during which patients are closely monitored for both bleeding events and potential thrombotic complications. This period is crucial to ensure that remission is stable and to assess whether emicizumab can be safely discontinued or if further treatment adjustments are necessary.
Emicizumab Safety
This systematic review indicates that emicizumab can be safely used in AHA patients, including those with cardiovascular risk factors. While thrombosis related to BPAs was notably highlighted in the EACH2 study, 7 the thrombotic risk associated with emicizumab appears to be low, though not fully defined. The GTH-AHA-EMI study reported fewer thromboembolic events compared to the GTH-AH01/2010 study, likely due to a decreased reliance on BPAs. 57 In fact, the combination of emicizumab with rFVIIa or rpFVIII39,45 has been reported efficious and safe from cumulative risk of thromboembolic complications while aPCC increased thrombotic microangiopathy. 66 However, an analysis of adverse events from the EudraVigilance pharmacovigilance database revealed 24 thromboembolic events in patients treated with emicizumab, with 25% occurring in combination with rFVIIa. 67 Some of these patients had AHA, and most complications were linked to multiple thromboembolism risk factors. Although the low rate of thromboembolism observed in the GTH-AHA-EMI study is promising, further investigation into this risk is needed.
In the absence of safety data, combining emicizumab with BPAs should be avoided in elderly patients with high cardiovascular risk. In cases where IST is effective, the combined effect of restored endogenous FVIII activity and ongoing emicizumab treatment may increase the risk of hypercoagulation. This transitional period requires careful monitoring and regular screening for signs of hypercoagulation.
Although the development of alloantibodies against emicizumab has been observed in individuals with CHA, it may also potentially occur in those with AHA. Two cases of anti-drug antibodies (ADAs) were detected only during the safety follow-up period after emicizumab administration. 22 Future monitoring is essential, as autoimmune conditions might increase the risk of developing ADAs. Further research is needed to explore any potential temporal link between the discontinuation of emicizumab treatment due to AHA remission and the emergence of ADAs.
Emicizumab Efficacy
While emicizumab reduces the frequency of bleeding in AHA patients, it does not entirely prevent breakthrough bleeding or even life-threatening hemorrhages. A few cases of rebleeding have been reported in patients taking emicizumab. This bleeding was mostly trigged by surgery,30,35,37,38 trauma injury23,26,36,38 or insufficient dose.26,46 For major surgeries under emicizumab, concurrent BPA with factor replacement is recommended. 68 rFVIIa and rpFVIII are preferred over aPCC for treating breakthrough bleeding due to concerns about thrombotic microangiopathy.Human FVIII concentrates may be used if the preferred agents are unavailable especially for patients with low-titer inhibitors. 58 Rehabilitation and minor surgeries can be safely performed under emicizumab prophylaxis. 22 Additionally, long-term emicizumab prophylaxis, lasting up to 1.75 years, has demonstrated sustained bleed prevention. 22
Further research is needed to address remaining questions about emicizumab, including its thrombotic risks when used with BPA and its safety during pregnancy, breastfeeding, and in pediatric populations. While emicizumab has been effective in pregnant women with congenital hemophilia, 56 its use in similar scenarios for AHA patients requires clinical trials to address these uncertainties.
Study Limitations
This systematic review has several limitations. First, the quality of evidence is limited, as most studies were case series, case reports, or conference abstracts with small sample sizes. Even the prospective AGEHA and GTH-AHA studies are constrained by limited sample sizes and the absence of a placebo control group, which may affect the analysis of results. Additionally, some published papers have limited data on patient follow-up.
Conclusion
The early use of emicizumab provides a novel approach to managing AHA, offering rapid and effective hemostatic control with significantly lower bleeding rates, thromboembolic events, and fatal infections, along with promising survival outcomes.Emicizumab is primarily intended for long-term prevention rather than acute bleeding episodes. It should be considered for AHA patients at diagnosis, with patient consent, though it is not yet licensed specifically for AHA. The adjusted dosing regimen over 2 days aims to quickly achieve hemostatic effectiveness and allows for deferral of IST for up to 12 weeks in ineligible patients. Despite minor risks of adverse events, emicizumab should be used with caution, especially in patients with high cardiovascular risk. Biological monitoring should be conducted regularly using chromogenic assays with bovine reagents until complete response and for several months thereafter. Due to the limited quality of current studies, further research is needed to assess the long-term safety of emicizumab, and establish a regular monitoring schedule to meet treatment completion criteria.
Acknowledgments
None
Footnotes
Authors contributions: All authors contributed to the conception and design of the study, the analysis and interpretation of data, the revision and the final approval of the version to be submitted.
Consent for publication: Not applicable
Consent to participate: Not applicable
Data availability statement: The data that support the finding ara available from the correspondang author upon reasonable request.
The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Ethical approval and informed consent statements: Not applicable
Funding: The authors received no financial support for the research, authorship, and/or publication of this article.
ORCID iD: Ghachem Ikbel https://orcid.org/0000-0003-3909-3868
References
- 1.Franchini M, Vaglio S, Marano G, et al. Acquired hemophilia A: A review of recent data and new therapeutic options. Hematology. 2017;22(9):514-520. doi: 10.1080/10245332.2017.1319115 [DOI] [PubMed] [Google Scholar]
- 2.Collins PW, Hirsch S, Baglin TP, et al. Acquired hemophilia A in the United Kingdom: A 2-year national surveillance study by the United Kingdom haemophilia centre Doctors’ organisation. Blood. 2007;109(5):1870-1877. doi: 10.1182/blood-2006-06-029850 [DOI] [PubMed] [Google Scholar]
- 3.Haider MZ, Anwer F. Acquired Hemophilia. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 [cité 13 mars 2023]: http://www.ncbi.nlm.nih.gov/books/NBK560494/.
- 4.Borg JY, Guillet B, Le Cam-Duchez V, et al. Outcome of acquired haemophilia in France: The prospective SACHA (surveillance des auto antiCorps au cours de l’Hémophilie acquise) registry. Haemophilia. 2013;19(4):564-570. doi: 10.1111/hae.12138 [DOI] [PubMed] [Google Scholar]
- 5.Salaj P, Geierová V, Ivanová E, et al. Identifying risk factors and optimizing standard of care for patients with acquired haemophilia A: Results from a Czech patient cohort. Haemophilia. 2020;26(4):643-651. DOI: 10.1111/hae.14084 [DOI] [PubMed] [Google Scholar]
- 6.Tiede A, Klamroth R, Scharf RE, et al. Prognostic factors for remission of and survival in acquired hemophilia A (AHA): Results from the GTH-AH 01/2010 study. Blood. 2015;125(7):1091-1097. doi: 10.1182/blood-2014-07-587089 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Knoebl P, Marco P, Baudo F, et al. Demographic and clinical data in acquired hemophilia A: Results from the European Acquired Haemophilia Registry (EACH2). J Thromb Haemostasis. 2012;10(4):622-631. doi: 10.1111/j.1538-7836.2012.04654.x [DOI] [PubMed] [Google Scholar]
- 8.Janbain M, Leissinger CA, Kruse-Jarres R. Acquired hemophilia A: Emerging treatment options. J Blood Med. 2015;6:143-150. doi: 10.2147/JBM.S77332 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Baudo F, Collins P, Huth-Kühne A, et al. Management of bleeding in acquired hemophilia A: Results from the European Acquired Haemophilia (EACH2) registry. Blood. 2012;120(1):39-46. doi: 10.1182/blood-2012-02-408930 [DOI] [PubMed] [Google Scholar]
- 10.Kruse-Jarres R, St-Louis J, Greist A, et al. Efficacy and safety of OBI-1, an antihaemophilic factor VIII (recombinant), porcine sequence, in subjects with acquired haemophilia A. Haemophilia. 2015;21(2):162-170. doi: 10.1111/hae.12627 [DOI] [PubMed] [Google Scholar]
- 11.Tiede A, Amano K, Ma A, et al. The use of recombinant activated factor VII in patients with acquired haemophilia. Blood Rev. 2015;29(Suppl 1):S19-S25. doi: 10.1016/S0268-960X(15)30004-7 [DOI] [PubMed] [Google Scholar]
- 12.Tiede A, Worster A. Lessons from a systematic literature review of the effectiveness of recombinant factor VIIa in acquired haemophilia. Ann Hematol. 2018;97(10):1889-1901. doi: 10.1007/s00277-018-3372-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Tiede A, Collins P, Knoebl P, et al. International recommendations on the diagnosis and treatment of acquired hemophilia A. Haematologica. 2020;105(7):1791-1801. doi: 10.3324/haematol.2019.230771 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Tiede A, Hofbauer CJ, Werwitzke S, et al. Anti-factor VIII IgA as a potential marker of poor prognosis in acquired hemophilia A: Results from the GTH-AH 01/2010 study. Blood. 2016;127(19):2289-2297. doi: 10.1182/blood-2015-09-672774 [DOI] [PubMed] [Google Scholar]
- 15.Sampei Z, Igawa T, Soeda T, et al. Identification and multidimensional optimization of an asymmetric bispecific IgG antibody mimicking the function of factor VIII cofactor activity. PLoS One. 2013;8(2):e57479. doi: 10.1371/journal.pone.0057479 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kitazawa T, Igawa T, Sampei Z, et al. A bispecific antibody to factors IXa and X restores factor VIII hemostatic activity in a hemophilia A model. Nat Med. 2012;18(10):1570-1574. doi: 10.1038/nm.2942 [DOI] [PubMed] [Google Scholar]
- 17.Holstein K, Albisetti M, Bidlingmaier C, et al. Practical guidance of the GTH haemophilia board on the use of emicizumab in patients with haemophilia A. Hamostaseologie. 2020;40(5):561-571. doi: 10.1055/a-1127-6476 [DOI] [PubMed] [Google Scholar]
- 18.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. doi: 10.1055/s-0039-1688687 [DOI] [PubMed] [Google Scholar]
- 19.Oldenburg J, Mahlangu JN, Kim B, et al. Emicizumab prophylaxis in hemophilia A with inhibitors. N Engl J Med. 2017;377(9):809-818. doi: 10.1056/NEJMoa1703068 [DOI] [PubMed] [Google Scholar]
- 20.Pipe SW, Shima M, Lehle M, et al. Efficacy, safety, and pharmacokinetics of emicizumab prophylaxis given every 4 weeks in people with haemophilia A (HAVEN 4): A multicentre, open-label, non-randomised phase 3 study. Lancet Haematol. 2019;6(6):e295-e305. doi: 10.1016/S2352-3026(19)30054-7 [DOI] [PubMed] [Google Scholar]
- 21.Reyes A, Révil C, Niggli M, et al. Efficacy of emicizumab prophylaxis versus factor VIII prophylaxis for treatment of hemophilia A without inhibitors: Network meta-analysis and sub-group analyses of the intra-patient comparison of the HAVEN 3 trial. Curr Med Res Opin. 2019;35(12):2079-2087. doi: 10.1080/03007995.2020.1744549 [DOI] [PubMed] [Google Scholar]
- 22.Shima M, Suzuki N, Nishikii H, et al. Final Analysis Results from the AGEHA Study: Emicizumab Prophylaxis for Acquired Hemophilia A with or without Immunosuppressive Therapy. Thromb Haemost. August 2024. doi: 10.1055/a-2384-3585. Thromb Haemost. 2024 Aug 12. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Shima M, Amano K, Ogawa Y, et al. 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. doi: 10.1016/j.jtha.2022.10.004 [DOI] [PubMed] [Google Scholar]
- 24.Tiede A, Hart C, Knöbl P, et al. Emicizumab prophylaxis in patients with acquired haemophilia A (GTH-AHA-EMI): An open-label, single-arm, multicentre, phase 2 study. Lancet Haematol. 2023;10(11):e913-e921. doi: 10.1016/S2352-3026(23)00280-6 [DOI] [PubMed] [Google Scholar]
- 25.Dane KE, Lindsley JP, Streiff MB, et al. Successful use of emicizumab in a patient with refractory acquired hemophilia A and acute coronary syndrome requiring percutaneous coronary intervention. Res Pract Thromb Haemost. 2019;3(3):420-423. doi: 10.1002/rth2.12201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Escobar M. Emicizumab prophylaxis in a patient with acquired hemophilia (Texas protocol): Case report. Haemophilia. 2021;27(S2):18-181. 10.1111/hae.14236 [DOI] [Google Scholar]
- 27.Escobar M, Montanez N. Prophylactic potential of standard and modified emicizumab prophylaxis in 2 patients with acquired hemophilia: A case report - ISTH congress abstracts. Res Pract Thromb Haemost. 2020;4(S1):385-387. [Google Scholar]
- 28.Knoebl P, Thaler J, Jilma P, et al. Emicizumab for the treatment of acquired hemophilia A. Blood. 2021;137(3):410-419. doi: 10.1182/blood.2020006315 [DOI] [PubMed] [Google Scholar]
- 29.Jena SS, Meher D, Dhankar N. Unforeseen encounter of acquired hemophilia A in a preoperative case of periampullary carcinoma: A case report. Int J Surg Case Rep. 2021;79:146-149. doi: 10.1016/j.ijscr.2021.01.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chen EC, Gibson WJ, Temoczko P, et al. Treatment of acquired hemophilia a with rituximab and emicizumab. Blood. 2020;136(Supplement 1):18-19. 10.1182/blood-2020-137262 [DOI] [Google Scholar]
- 31.Al-Banaa K, Alhillan A, Hawa F, et al. Emicizumab use in treatment of acquired hemophilia A: A case report. Am J Case Rep. 2019;20:1046-1048. doi: 10.12659/AJCR.916783 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Ganslmeier M, Pekrul I, Heinrich DA, et al. Persistent inhibitor in acquired haemophilia A: A case for emicizumab? Haemophilia. 2021;27(4):e502-e505. doi: 10.1111/hae.14225 [DOI] [PubMed] [Google Scholar]
- 33.Hess KJ, Patel P, Joshi AM, et al. Utilization of Emicizumab in Acquired Factor VIII Deficiency. Am J Case Rep. 2020;21. doi: 10.12659/AJCR.922326 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Möhnle P, Pekrul I, Spannagl M, et al. Emicizumab in the treatment of acquired haemophilia: A case report. Transfus Med Hemother. 2019;46(2):121-123. doi: 10.1159/000497287 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Flommersfled S, Sachs U. Successful control of bleeding with emicizumab in aquired haemophilia A: A case report. Hamostaseologie. 2019;39(s1):1-92. DOI : 10.1055/S-0039-168016630776836 [Google Scholar]
- 36.Chen S-L, Ellsworth P, Kasthuri RS, et al. Emicizumab reduces re-hospitalization for bleeding in acquired haemophilia A. Haemophilia. 2021;27(4):e585-e588. doi: 10.1111/hae.14335 [DOI] [PubMed] [Google Scholar]
- 37.Chen EC, Gibson W, Temoczko P, et al. Emicizumab for the treatment of acquired hemophilia A: Retrospective review of a single-institution experience. Haemophilia. September 2022;29(1):84-89. doi: 10.1111/hae.14664 [DOI] [PubMed] [Google Scholar]
- 38.Poston JN, Al-Banaa K, von Drygalski A, et al. Emicizumab for the treatment of acquired hemophilia a: A multicenter US case series. Blood. 2021;138:496. doi: 10.1182/blood-2021-148040 [DOI] [Google Scholar]
- 39.Happaerts M, Vanassche T. Acquired hemophilia following COVID-19 vaccination: Case report and review of literature. Research and Practice in Thrombosis and Haemostasis. 2022;6(6):e12785. doi: 10.1002/rth2.12785 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Karthick RG, Prabhu S, Ram B, et al. Acquired hemophilia-treated with rituximaband emicizumab-A case report. Indian Journal of Hematology and Blood Transfusion. 2021:57. doi: 10.1007/s12288-021-01510-0 [DOI] [Google Scholar]
- 41.Nishikii H, Suzuki N, Kobayashi R, et al. Efficacy and safety of emicizumab prophylaxis in patients with acquired hemophilia a who were deemed ineligible for immunosuppressive therapy: Additional data from the ageha study. Blood. 2022;140(Supplement 1):2724-2725. 10.1182/blood-2022-162237 [DOI] [Google Scholar]
- 42.Hansenne A, Hermans C. Emicizumab in acquired haemophilia A: About two clinical cases and literature review. Ther Adv Hematol. 2021;12:20406207211038193. doi: 10.1177/20406207211038193 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hayden A, Candelario N, Moyer G. Recombinant porcine factor VIII in acquired hemophilia A: Experience from two patients and literature review. Res Pract Thromb Haemost. 2022;6(2):e12688. doi: 10.1002/rth2.12688 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Gelbbenger G, Ludwing T, Knoebl P. Management of acquired haemophilia A in severe COVID-19: Haemostatic bridging with emicizumab to keep the balance between bleeding and thrombosis. Br J Clin Pharmacol. 2023 Feb;89(2):908-913. doi: 10.1111/bcp.15598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kaunchale N, Ansari S, More A, et al. Successful treatment of acquired hemophilia A using FEIBA supplemented with emicizumab. Indian J Hematol Blood Transfus. 2023 Jan;39(1):159-160. doi: 10.1007/s12288-022-01566-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yates SG, Webb CB, Sarode R, et al. Utilization of emicizumab in acquired hemophilia A: A case report. Transfus Apher Sci. 2022;61(6):103457. doi: 10.1016/j.transci.2022.103457 [DOI] [PubMed] [Google Scholar]
- 47.Knöbl P, Thaler J, Jilma P, Quehenberger P, Gleixner K, Sperr W. Emicizumab for the treatment of acquired hemophilia A: an update of the Vienna experience [Internet]. ISTH Congress Abstracts: https://abstracts.isth.org/abstract/emicizumab-for-the-treatment-of-acquired-hemophilia-a-an-update-of-the-vienna-experience/. [DOI] [PubMed]
- 48.Latef TJ, Bhardwaj P, Bilal M. Refractory acquired haemophilia A in a patient with HIV treated with emicizumab. Blood Coagul Fibrinolysis. 2022;33(2):138. doi: 10.1097/MBC.0000000000001118 [DOI] [PubMed] [Google Scholar]
- 49.Crossette-Thambiah C, Arachchillage D, Laffan M. Post-partum Acquired Haemophilia A in the COVID era – building the case for Emicizumab? [Internet]. ISTH Congress Abstracts: https://abstracts.isth.org/abstract/post-partum-acquired-haemophilia-a-in-the-covid-era-building-the-case-for-emicizumab/.
- 50.Al-Banaa K, Gallastegui-Crestani N, von Drygalski A. Anticoagulation for stroke prevention after restoration of haemostasis with emicizumab in acquired haemophilia A. Eur J Case Rep Intern Med. 2021;8(11):002984. doi: 10.12890/2021_002984 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Engelen MM, Vandesande J, De Bent J, et al. Emicizumab for acquired haemophilia A: A case series. Haemophilia. 2023;29(4):1049-1055. doi: 10.1111/hae.14809 [DOI] [PubMed] [Google Scholar]
- 52.Ahmed F, Kasianchyk M, Moreno A, et al. Emicizumab for acquired hemophilia A: Report of two cases and dosing strategies. EJHaem. 2024;5(2):387-391. doi: 10.1002/jha2.878 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Pipe SW, Collins P, Dhalluin C, et al. Emicizumab prophylaxis in infants with hemophilia A (HAVEN 7): Primary analysis of a phase 3b open-label trial. Blood. 2024;143(14):1355-1364. 10.1182/blood.2023021832 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Schmitt C, Adamkewicz JI, Xu J, et al. Pharmacokinetics and pharmacodynamics of Emicizumab in persons with hemophilia A with factor VIII inhibitors: HAVEN 1 study. Thromb Haemost. 2021;121(3):351-360. doi: 10.1055/s-0040-1717114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Nakajima Y, Takami E, Nakano H, et al. In vitro evaluation of global coagulation potentials in the co-presence of plasma-derived factors viia/X products (byclot®) and emicizumab in patients with haemophilia A and inhibitors and acquired haemophilia A: A pilot study. Haemophilia. 2022;28(5):e149-e152. doi: 10.1111/hae.14650 [DOI] [PubMed] [Google Scholar]
- 56.Mahlangu J, Oldenburg J, Paz-Priel I, et al. Emicizumab prophylaxis in patients who have hemophilia A without inhibitors. N Engl J Med. 2018;379(9):811-822. doi: 10.1056/NEJMoa1803550 [DOI] [PubMed] [Google Scholar]
- 57.Hart C, Klamroth R, Sachs UJ, et al. Emicizumab versus immunosuppressive therapy for the management of acquired hemophilia A. J Thromb Haemost. June 2024;S1538-7836(24):00368‐4. doi: 10.1016/j.jtha.2024.06.010 [DOI] [PubMed] [Google Scholar]
- 58.Pfrepper C, Klamroth R, Oldenburg J, et al. Emicizumab for the Treatment of Acquired Hemophilia A: Consensus Recommendations from the GTH-AHA Working Group. Hamostaseologie. December 2023. doi: 10.1055/a-2197-9738 [DOI] [PubMed] [Google Scholar]
- 59.Takeyama M, Nogami K, Matsumoto T, et al. An anti-factor IXa/factor X bispecific antibody, emicizumab, improves ex vivo coagulant potentials in plasma from patients with acquired hemophilia A. J Thromb Haemost. 2020;18(4):825-833. doi: 10.1111/jth.14746 [DOI] [PubMed] [Google Scholar]
- 60.Bansal S, Donners AAMT, Fischer K, et al. Low dose emicizumab prophylaxis in haemophilia a patients: A pilot study from India. Haemophilia. 2023;29(3):931-934. doi: 10.1111/hae.14785 [DOI] [PubMed] [Google Scholar]
- 61.Chuansumrit A, Sirachainan N, Jaovisidha S, et al. Effectiveness of monthly low dose emicizumab prophylaxis without 4-week loading doses among patients with haemophilia A with and without inhibitors: A case series report. Haemophilia. 2023;29(1):382-385. doi: 10.1111/hae.14707 [DOI] [PubMed] [Google Scholar]
- 62.Dobbelstein C, Moschovakis GL, Tiede A. Reduced-intensity, risk factor–stratified immunosuppression for acquired hemophilia A: Single-center observational study. Ann Hematol. 2020;99(9):2105-2112. doi: 10.1007/s00277-020-04150-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Schutgens REG, Jimenez-Yuste V, Escobar M, et al. Antithrombotic treatment in patients with hemophilia: An EHA-ISTH-EAHAD-ESO clinical practice guidance. Hemasphere. 2023;7(6):e900. doi: 10.1097/HS9.0000000000000900 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Peyvandi F, Kenet G, Pekrul I, et al. Laboratory testing in hemophilia: Impact of factor and non-factor replacement therapy on coagulation assays. J Thromb Haemost. 2020;18(6):1242-1255. doi: 10.1111/jth.14784 [DOI] [PubMed] [Google Scholar]
- 65.Miller CH, Boylan B, Payne AB, et al. Validation of the chromogenic Bethesda assay for factor VIII inhibitors in hemophilia a patients receiving Emicizumab. Int J Lab Hematol. 2021;43(2):e84-e86. doi: 10.1111/ijlh.13384 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tiede A, Kemkes-Matthes B, Knöbl P. Should emicizumab be used in patients with acquired hemophilia A? J Thromb Haemostasis. 2021;19(3):637-644. doi: 10.1111/jth.15208 [DOI] [PubMed] [Google Scholar]
- 67.Abbattista M, Ciavarella A, Noone D, et al. Hemorrhagic and thrombotic adverse events associated with emicizumab and extended half-life factor VIII replacement drugs: EudraVigilance data of 2021. J Thromb Haemost. 2023;21(3):546-552. doi: 10.1016/j.jtha.2023.01.010 [DOI] [PubMed] [Google Scholar]
- 68.Castaman G, Peyvandi F, Kremer Hovinga JA, et al. Surgical experience from the STASEY study of emicizumab prophylaxis in people with hemophilia A with factor VIII inhibitors. TH Open. 2024;8(1):e42-e54. doi: 10.1055/s-0043-1777766 [DOI] [PMC free article] [PubMed] [Google Scholar]

