A 77-year-old man with atrial fibrillation and a CHA2DS2Vasc score of 6 for hypertension, age, diabetes, and previous stroke is brought to the emergency department with decreased level of consciousness. He is anticoagulated with rivaroxaban (a direct oral factor Xa inhibitor [FXaI]) and received his last dose about 4 hours before presentation. Urgent computed tomography of the head shows intracerebral hemorrhage. Because of his previous stroke, the patient’s family is concerned about treating the bleed with pharmacological agents that may increase the risk of stroke. What are the risks of thrombosis and mortality related to the use of prothrombin complex concentrates (PCCs) and andexanet alfa for patients with direct oral FXaI-associated major bleeding?

Learning Objectives

  • Summarize the evidence regarding the use of prothrombin complex concentrates and andexanet alfa for the management of direct oral factor Xa inhibitor–associated major bleeding

  • Calculate the rates and cumulative incidences of mortality and thrombosis related to the use of prothrombin complex concentrates and andexanet alfa for the management of direct oral factor Xa inhibitor–associated major bleeding

Direct oral anticoagulants are widely used to prevent and treat thromboembolism, most often to reduce the risk of stroke and embolic phenomena in atrial fibrillation and for the treatment and secondary thromboprophylaxis of venous thromboembolism. Idarucizumab, a specific reversal agent for the direct oral thrombin inhibitor dabigatran, is approved for the management of major bleeding events (MBEs) or in preparation for urgent surgery. Andexanet alfa was designed as a specific reversal agent for direct and indirect FXaIs and is currently approved in the United States and Europe for the reversal of apixaban and rivaroxaban in patients with MBE on the basis of the registration study (Prospective, Open-Label Study of Andexanet Alfa in Patients Receiving a Factor Xa Inhibitor Who Have Acute Major Bleeding [ANNEXA-4]). Andexanet is a recombinant, inactive form of factor Xa that binds and sequesters the FXaI and therefore neutralizes its anticoagulant effect. However, andexanet is not widely available, and its use is associated with important cost implications for hospitals and health care systems. PCCs have been used off-label for the management of MBE in patients receiving direct oral FXaIs. Four-factor PCC (4F-PCC) contains the vitamin K–dependent coagulation factors II, VII, IX, and X, whereas the 3-factor PCC does not contain factor VII. Some centers have developed protocols with activated prothrombin complex concentrates (aPCCs) for the management of FXaI-associated MBE. aPCC contains activated factor VII and mostly inactivated factors II, IX, and X. A recent systematic review aimed to characterize the hemostatic efficacy of PCC for the management of FXaI-associated MBE.1  However, a meta-analysis of the hemostatic efficacy of PCC for this indication could not be performed, owing to the heterogeneous definitions of adequate hemostasis adopted by the included studies. Therefore, we sought to summarize the safety outcomes of these FXaI-associated MBE management strategies by providing early (in-hospital) and late (at 30-day follow-up) rates and cumulative incidences for overall mortality and thrombotic complications.

We systematically searched MEDLINE and Embase to identify articles assessing mortality and thrombotic complications associated with the use of PCC, aPCC, and andexanet for the management of FXaI-associated major bleeding. We also sought to explore the different definitions of hemostatic effectiveness used in studies assessing these different hemostatic agents for the management of MBE in patients receiving FXaI. We modified and updated a search strategy devised for the synthesis of evidence in the context of the American Society of Hematology guideline development on optimal management of anticoagulation therapy.1  We included published randomized controlled trials, prospective cohort studies, retrospective cohort studies, and case series (with at least 10 patients) designed to evaluated the safety and efficacy of PCC, aPCC, or andexanet for the management of FXaI-associated MBE. Studies of patients needing hemostatic support in preparation for emergent surgery were excluded because the objective of the review was to inform clinicians regarding the risks of death and thrombosis in patients receiving PCC, aPCC, or andexanet for FXaI-associated MBE. Title and abstract screening, full-text review, and data extraction were conducted in duplicate by 2 independent reviewers using Covidence software (Covidence, Melbourne, Australia). When appropriate, pooled rates and cumulative incidences for overall mortality and thrombosis were calculated using StatsDirect software (StatsDirect, Birkenhead, UK).

A total of 1459 citations were identified (Figure 1). After duplicate removal, 1282 references were screened. There were 146 references that underwent full-text review, and 15 were included. Of those, 13 observational studies assessed the use of PCC, 1 retrospective study examined the use of aPCC, and 1 prospective study addressed the use of andexanet in our patient population. The majority of excluded studies at the full-text review stage were abstracts, and a smaller number did not contain the appropriate patient population or report outcomes of interest or did not have the included study designs. The risk of bias of included studies was assessed according to guidance for nonrandomized studies provided in the Cochrane Handbook.2 

Figure 1.

Study flow diagram. AA, andexanet alfa; aPCC, activated prothrombin complex concentrate; PCC, prothrombin complex concentrate.

Figure 1.

Study flow diagram. AA, andexanet alfa; aPCC, activated prothrombin complex concentrate; PCC, prothrombin complex concentrate.

Close modal

PCCs

Thirteen nonrandomized, unblinded studies including 521 patients evaluated the use of PCCs in the management of MBE in patients receiving an FXaI (Table 1). Four studies were prospective,3-6  and nine studies were retrospective.7-15  The majority (69%) of studies followed patients only during hospital admission.6,7,9-15  On average, the hospital admission lasted 6.6 days. Four studies had a follow-up period of 30 days.3-5,8  Most of the included patients were receiving rivaroxaban for stroke prophylaxis in atrial fibrillation. Intracranial hemorrhage was the most common reason for the administration of PCC. A minority (38%) of studies defined major bleeding per the International Society on Thrombosis and Haemostasis (ISTH) definition.16  The definition of effective hemostasis was provided in 9 studies and varied across studies, with 2 studies3,14  using the ISTH definition,17  2 studies5,15  using the definition proposed by Sarode et al,18  and 5 studies4,7-9,11  using their own definition of effective hemostasis. Most patients were judged to have achieved adequate hemostasis on the basis of various definitions of hemostatic effectiveness employed by the included studies. All studies were judged to be at high risk for bias as single-cohort studies lacking control groups.

Table 1.

Characteristics of the 13 included studies assessing the use of prothrombin complex concentrate in factor Xa inhibitor–associated major bleeding

ReferenceStudy designNo.TreatmentMajor bleeding definitionFactor Xa inhibitorIndication for anticoagulationFollow-up (d)Intracranial hemorrhageGastrointestinal bleedingOther bleeding
Allison7  Retrospective 33 4F-PCC (35 U/kg) Other Apixaban = 6 AF = 24 7.1 30 
Rivaroxaban = 27 VTE = 6 
 Other = 3 
Arachchillage8  Retrospective 80 4F-PCC ISTH Apixaban = 40 AF = 67 30 46 24 10 
Rivaroxaban = 40 VTE = 12 
 Both = 1 
Berger9  Retrospective 22 4F-PCC (25 U/kg) Other Apixaban = 5 AF = 17 5.9 22 — — 
Rivaroxaban = 15 VTE = 4 
Dabigatran = 2 Other = 3 
 (>1 possible per patient) 
Dybdahl10  Retrospective 35 4F-PCC (50 U/kg) Other Apixaban = 17 AF = 30 6.4 35 — — 
Rivaroxaban = 18 VTE = 4 
 Both = 1 
Grandhi11  Retrospective 18 4F-PCC Other Apixaban = 2 AF = 15 8.4 18 — — 
Rivaroxaban = 18 VTE = 1 
 Both = 1 
 Other = 1 
Harrison12  Retrospective 14 4F-PCC Other N/A AF = 12 7.1 14 — — 
VTE = 3 
Other = 2 
(>1 possible per patient) 
Majeed3  Prospective 84 4F-PCC (25 U/kg) ISTH Apixaban = 39 AF = 63 30 59 13 12 
Rivaroxaban = 45 VTE = 3 
 Both = 18 
Müller13  Retrospective 74* 4F-PCC N/A Apixaban = 5 AF = 48 4.9 45 14 14 
Rivaroxaban = 67 VTE = 13 
Edoxaban = 1 Other = 13 
Dabigatran = 1  
Schenk4  Prospective 13 4F-PCC (25 U/kg) N/A Rivaroxaban = 13 N/A 30 10 
Schulman5  Prospective 66 4F-PCC (2000 U) ISTH Apixaban = 29 AF = 54 30 36 16 14 
Rivaroxaban = 37 VTE = 8 
 Both = 2 
 Other = 1 
Sheikh-Taha14  Retrospective 29 4F-PCC (50 U/kg) ISTH Apixaban = 13 AF = 23 5.9 21 
Rivaroxaban = 16 VTE = 5 
 Other = 1 
Smith15  Retrospective 31 4F-PCC (25-50 U/kg) Other Apixaban = 17 AF = 28 7.2 18 12 
Rivaroxaban = 14 VTE = 3 
Testa6  Prospective 22 PCC ISTH Apixaban = 6 N/A 20 
Rivaroxaban = 16 
ReferenceStudy designNo.TreatmentMajor bleeding definitionFactor Xa inhibitorIndication for anticoagulationFollow-up (d)Intracranial hemorrhageGastrointestinal bleedingOther bleeding
Allison7  Retrospective 33 4F-PCC (35 U/kg) Other Apixaban = 6 AF = 24 7.1 30 
Rivaroxaban = 27 VTE = 6 
 Other = 3 
Arachchillage8  Retrospective 80 4F-PCC ISTH Apixaban = 40 AF = 67 30 46 24 10 
Rivaroxaban = 40 VTE = 12 
 Both = 1 
Berger9  Retrospective 22 4F-PCC (25 U/kg) Other Apixaban = 5 AF = 17 5.9 22 — — 
Rivaroxaban = 15 VTE = 4 
Dabigatran = 2 Other = 3 
 (>1 possible per patient) 
Dybdahl10  Retrospective 35 4F-PCC (50 U/kg) Other Apixaban = 17 AF = 30 6.4 35 — — 
Rivaroxaban = 18 VTE = 4 
 Both = 1 
Grandhi11  Retrospective 18 4F-PCC Other Apixaban = 2 AF = 15 8.4 18 — — 
Rivaroxaban = 18 VTE = 1 
 Both = 1 
 Other = 1 
Harrison12  Retrospective 14 4F-PCC Other N/A AF = 12 7.1 14 — — 
VTE = 3 
Other = 2 
(>1 possible per patient) 
Majeed3  Prospective 84 4F-PCC (25 U/kg) ISTH Apixaban = 39 AF = 63 30 59 13 12 
Rivaroxaban = 45 VTE = 3 
 Both = 18 
Müller13  Retrospective 74* 4F-PCC N/A Apixaban = 5 AF = 48 4.9 45 14 14 
Rivaroxaban = 67 VTE = 13 
Edoxaban = 1 Other = 13 
Dabigatran = 1  
Schenk4  Prospective 13 4F-PCC (25 U/kg) N/A Rivaroxaban = 13 N/A 30 10 
Schulman5  Prospective 66 4F-PCC (2000 U) ISTH Apixaban = 29 AF = 54 30 36 16 14 
Rivaroxaban = 37 VTE = 8 
 Both = 2 
 Other = 1 
Sheikh-Taha14  Retrospective 29 4F-PCC (50 U/kg) ISTH Apixaban = 13 AF = 23 5.9 21 
Rivaroxaban = 16 VTE = 5 
 Other = 1 
Smith15  Retrospective 31 4F-PCC (25-50 U/kg) Other Apixaban = 17 AF = 28 7.2 18 12 
Rivaroxaban = 14 VTE = 3 
Testa6  Prospective 22 PCC ISTH Apixaban = 6 N/A 20 
Rivaroxaban = 16 

4F, four-factor; AF, atrial fibrillation; ISTH, International Society on Thrombosis and Haemostasis; N/A, not available; PCC, prothrombin complex concentrate; VTE, venous thromboembolism .

*

93% of patients received PCCs for major bleeding.

Using a random effects model, the pooled in-hospital overall mortality rate was 3.4 (95% confidence interval [CI], 2.5-4.3; I2 = 2.7%) per 100 person-days, and the pooled in-hospital cumulative overall mortality was 18% (95% CI, 14% to 23%; I2 = 2.6%) (Table 2). The pooled in-hospital thrombosis rate was 0.6 (95% CI, 0.2-1.1; I2 = 4.8%) per 100 person-days, and the pooled in-hospital cumulative incidence of thrombotic events was 3% (95% CI, 1% to 7%; I2 = 0%). The pooled 30-day overall mortality rate was 1.0 (95% CI, 0.6-1.6; I2 = 69.9%) per 100 person-days, and the pooled 30-day cumulative overall mortality was 26% (95% CI, 16% to 38%; I2 = 69.6%) (Table 3). The pooled 30-day thrombotic complication rate was 0.2 (95% CI, 0.1-0.4; I2 = 0%) per 100 person-days, and the pooled 30-day cumulative incidence of thrombotic events was 6% (95% CI, 3% to 9%; I2 = 13.4%).

Table 2.

In-hospital mortality and thrombotic complications in patients treated with prothrombin complex concentrate for a factor Xa inhibitor–associated major bleeding event

ReferenceOverall in-hospital mortality, nOverall in-hospital cumulative mortality, %Overall in-hospital mortality rate (per 100 person-days)In-hospital thrombotic events, nIn-hospital thrombotic events cumulative incidence, %In-hospital thrombotic event rate (per 100 person-days)Hemostatic adequacy*, n (%)
Allison7  15% 2.5 26 (84%) 
Berger9  18% 3.8 9% 1.9 17 (94%) 
Dybdahl10  23% 4.0 3% 0.5 N/A 
Grandhi11  33% 4.8 6% 0.8 17 (94%) 
Harrison12  14% 2.2 13 (93%) 
Müller13  9% 2.0 N/A N/A N/A N/A 
Sheikh-Taha14  21% 3.9 3% 0.7 21 (72%) 
Smith15  16% 2.4 25 (81%) 
Testa6  23% 8.5 N/A N/A N/A N/A 
ReferenceOverall in-hospital mortality, nOverall in-hospital cumulative mortality, %Overall in-hospital mortality rate (per 100 person-days)In-hospital thrombotic events, nIn-hospital thrombotic events cumulative incidence, %In-hospital thrombotic event rate (per 100 person-days)Hemostatic adequacy*, n (%)
Allison7  15% 2.5 26 (84%) 
Berger9  18% 3.8 9% 1.9 17 (94%) 
Dybdahl10  23% 4.0 3% 0.5 N/A 
Grandhi11  33% 4.8 6% 0.8 17 (94%) 
Harrison12  14% 2.2 13 (93%) 
Müller13  9% 2.0 N/A N/A N/A N/A 
Sheikh-Taha14  21% 3.9 3% 0.7 21 (72%) 
Smith15  16% 2.4 25 (81%) 
Testa6  23% 8.5 N/A N/A N/A N/A 

N/A, not available.

*

Based on evaluable patients, which may be a smaller sample than the total sample.

Table 3.

Thirty-day mortality and thrombotic complications in patients treated with prothrombin complex concentrate for a factor Xa inhibitor–associated major bleeding event

ReferenceOverall 30-d mortality, nOverall 30-d cumulative mortality, %Overall 30-d mortality rate (per 100 person-days)30-d thrombotic events, n30-d thrombotic events cumulative incidence, %30-d thrombotic event rate (per 100 person-days)Hemostatic adequacy, n (%)
Arachchillage8  27 34% 1.4 3.8% 0.2 59 (74%) 
Majeed3  27 32% 1.3 3.6% 0.1 58 (69%) 
Schenk4  23% 0.9 15.4% 0.6 10 (78%) 
Schulman5  14% 0.5 7.6% 0.3 43 (65%) 
ReferenceOverall 30-d mortality, nOverall 30-d cumulative mortality, %Overall 30-d mortality rate (per 100 person-days)30-d thrombotic events, n30-d thrombotic events cumulative incidence, %30-d thrombotic event rate (per 100 person-days)Hemostatic adequacy, n (%)
Arachchillage8  27 34% 1.4 3.8% 0.2 59 (74%) 
Majeed3  27 32% 1.3 3.6% 0.1 58 (69%) 
Schenk4  23% 0.9 15.4% 0.6 10 (78%) 
Schulman5  14% 0.5 7.6% 0.3 43 (65%) 

Activated prothrombin complex concentrate

One retrospective chart review evaluated aPCC in 3 patients with major bleeding receiving apixaban and 8 receiving rivaroxaban.19  All patients were receiving an FXaI for atrial fibrillation, and 2 patients had a concomitant diagnosis of venous thromboembolism. Only patients presenting with an intracranial bleeding event documented via computed tomographic scans were included. The patients were treated with factor VIII inhibitor bypassing agent (20 U/kg). Progression of major bleeding was assessed via repeated computed tomography in 91% of patients between 8 and 24 hours after initial imaging. Of these, 40% had worsening intracranial hemorrhage on repeat imaging. Overall, 27% died before discharge from the hospital, and 18% developed a thrombotic complication during their admission. It was not possible to provide a rate of all-cause mortality and thrombosis, because the length of follow-up for the cohort was not provided.

Andexanet alfa

ANNEXA-4 was a multicenter, prospective, open-label, single-arm study that evaluated andexanet alfa for patients receiving apixaban, edoxaban, rivaroxaban, or enoxaparin who presented with major bleeding.20  Major bleeding was defined as potentially life-threatening bleeding associated with hemodynamic compromise, a decrease in hemoglobin of ≥2 g/dL, or bleeding in a critical area or organ. Of note, patients were excluded if they were presenting with an intracranial hemorrhage and a Glasgow Coma Scale score <7, a hematoma volume estimated to be >60 mL, an expected survival <1 month, or thrombosis within the previous 2 weeks. The study had 2 coprimary endpoints: the percentage change from baseline in anti–factor Xa activity and the percentage of patients deemed to have good or excellent hemostasis 12 hours after andexanet infusion. The study outcomes were adjudicated independently by a committee. The safety analysis was comprised of all patients who received a dose of andexanet, including 194 patients receiving apixaban, 128 receiving rivaroxaban, and 10 receiving edoxaban. A small percentage (6%) of patients were receiving enoxaparin. Most patients were prescribed an anticoagulant for atrial fibrillation (80%). Patients presented with intracranial hemorrhage (65%), gastrointestinal bleeding (26%), or other (10%). During the 30-day follow-up, the overall mortality rate was 0.5 per 100 person-days, and the cumulative overall mortality was 14%. The 30-day thrombotic event rate was 0.4 per 100 person-days, and the cumulative incidence of thrombotic events was 10%. Most of the thrombotic events occurred before anticoagulation was restarted. Seventy-two percent of patients were deemed by independent adjudicators to have good or excellent hemostatic efficacy 12 hours after the infusion of the study drug, using criteria adapted from the Sarode et al study.18  Notably, compared with the criteria proposed by the ISTH,17  the criteria for effective hemostasis in intracranial hemorrhage were based solely on imaging results and did not include clinical deterioration or the need for repeat hemostatic products 48 hours after initial management.

This review highlights the methodologically limited evidence regarding the thrombosis and mortality risks associated with use of PCC, aPCC, and andexanet alfa for the management of FXaI-associated MBE. All studies were observational in nature, with no control groups for comparison, and therefore are at risk of selection bias with respect to included patients. Studies with retrospective data collection are also at risk of detection bias regarding outcome ascertainment and adjudication such that the true estimates of thrombosis are uncertain. Furthermore, there is insufficient evidence regarding the use of PCC or andexanet for patients treated with edoxaban or betrixaban.

Given these limitations, the pooled data on the use of PCC for the management of FXaI-associated MBE showed that 18% of patients died and 3% experienced a thrombotic complication during hospital admission. At 30 days after FXaI-associated MBE, 26% of patients had died and 6% had experienced thrombosis. The majority of the deaths occurred early after the MBE. In the 2 prospective studies of consecutive patients with centrally adjudicated outcomes, the majority of the deaths were attributed to the index bleeding event.3,5  The 30-day pooled mortality rate and cumulative incidence calculated for PCC were higher than those for andexanet, which may be due to baseline differences in patient populations. Furthermore, a substantial proportion of thrombotic events occurred after hospital discharge, suggesting that additional considerations for the development of thrombosis likely contribute to this risk, including withdrawal of anticoagulants in patients at high baseline risk for thromboembolism. In the 2 above-mentioned prospective studies, 78% of the thrombotic complications occurred before resumption of full-dose anticoagulation. These findings are consistent with those of the ANNEXA-4 study, in which half of the thrombotic events occurred beyond 5 days after the andexanet infusion, although the 30-day rate and cumulative incidence were found to be higher in the ANNEXA-4 study. This increase in thrombotic events may be related to differences in study design, particularly as they pertain to the detection of thrombotic events within a prespecified, standardized prospective follow-up period. The higher mortality rate seen in the PCC studies may also have led to a lower thrombotic event rate because patients who died would not have developed thrombosis. Of note, the cumulative incidence of thrombosis after 4F-PCC presented here is higher than that found in patients receiving warfarin, which was reported to be 1.8% in a previous meta-analysis (95% CI, 1.0–3.0).21  In 2 recent retrospective studies comparing PCC for the management of MBE in patients receiving warfarin and FXaI, however, the incidence of death and thromboembolic events did not differ significantly between the 2 groups.8,12  Therefore, the incremental contribution of PCCs and andexanet to the thrombotic risk in this setting remains uncertain.

This review has several limitations. Most of the studies were of small sample size, retrospective in nature, and at high risk of bias. Furthermore, an important selection bias was present in most of the included studies, in which the sickest patients were likely chosen to receive off-label hemostatic support agents, whereas patients with less critical bleeding or with other management options (such as local hemostatic measures) may not have been offered these interventions.

In summary, the quality of evidence for the use of PCC in FXaI-associated MBE is very low. In the event of a life-threatening bleed, it is reasonable to offer PCC for hemostatic support as an adjunct to other interventions, including FXaI cessation, promotion of adequate renal function, transfusion support, and procedural or surgical interventions to achieve local hemostasis. Evidence for aPCC is even more limited, precluding recommendations about its use in this setting. Andexanet is the only agent currently approved in the United States and Europe for management of apixaban- and rivaroxaban-associated major bleeds. However, its high cost and lack of availability remain significant challenges for its use in everyday practice. Further studies designed to randomize patients to a treatment or control group are needed to help better characterize the efficacy and safety of these interventions in the management of FXaI-MBE. The ongoing ANNEXA-I (A Phase 4 Randomized Clinical Trial of Andexanet Alfa [Andexanet Alfa for Injection] in Acute Intracranial Hemorrhage in Patients Receiving an Oral Factor Xa Inhibitor) study is evaluating andexanet compared with usual care in patients with acute intracranial hemorrhage receiving a direct oral FXaI (NCT03661528).

Recommendations

Andexanet alfa should be used for the management of apixaban- or rivaroxaban-associated intracranial or gastrointestinal major bleeding (Grading of Recommendations Assessment, Development, and Evaluation grade 2C).

4F-PCC should be administered as a hemostatic therapy for the management of apixaban-, rivaroxaban-, edoxaban-, or betrixaban-associated major bleeding (grade 2C).

There is insufficient evidence to preferentially recommend andexanet alfa or 4F-PCC on the basis of thrombosis or mortality risks (grade 2C).

There is insufficient evidence to recommend the use of aPCCs in the management of FXaI-associated major bleeding (grade 2C).

Physicians, patients, and caregivers should be advised about the uncertainty of efficacy and the risks of thrombosis and mortality associated with these 4F-PCCs and andexanet alfa for FXaI-associated major bleeding (grade 2C).

M.K. has received grant funding from the CanVECTOR network for her fellowship in adult thrombosis medicine. D.M.S. is supported by a research early career award from the Hamilton Health Sciences Foundation.

Deborah M Siegal, Population Health Research Institute, 20 Copeland Ave, C3-120, Hamilton, ON L8L 2X2, Canada; e-mail: siegald@mcmaster.ca.

1.
Piran
S
,
Khatib
R
,
Schulman
S
, et al
.
Management of direct factor Xa inhibitor-related major bleeding with prothrombin complex concentrate: a meta-analysis
.
Blood Adv
.
2019
;
3
(
2
):
158
-
167
.
2.
Higgins
JP
,
Green
S
, eds.
Cochrane Handbook for Systematic Reviews of Interventions
.
Chichester, United Kingdom
:
John Wiley & Sons Ltd
;
2008
.
3.
Majeed
A
,
Ågren
A
,
Holmström
M
, et al
.
Management of rivaroxaban- or apixaban-associated major bleeding with prothrombin complex concentrates: a cohort study
.
Blood
.
2017
;
130
(
15
):
1706
-
1712
.
4.
Schenk
B
,
Goerke
S
,
Beer
R
,
Helbok
R
,
Fries
D
,
Bachler
M
.
Four-factor prothrombin complex concentrate improves thrombin generation and prothrombin time in patients with bleeding complications related to rivaroxaban: a single-center pilot trial
.
Thromb J
.
2018
;
16
:
1
.
5.
Schulman
S
,
Gross
PL
,
Ritchie
B
, et al
;
Study Investigators
.
Prothrombin complex concentrate for major bleeding on factor Xa inhibitors: a prospective cohort study
.
Thromb Haemost
.
2018
;
118
(
5
):
842
-
851
.
6.
Testa
S
,
Ageno
W
,
Antonucci
E
, et al
.
Management of major bleeding and outcomes in patients treated with direct oral anticoagulants: results from the START-Event registry
.
Intern Emerg Med
.
2018
;
13
(
7
):
1051
-
1058
.
7.
Allison
TA
,
Lin
PJ
,
Gass
JA
, et al
.
Evaluation of the use of low-dose 4-factor prothrombin complex concentrate in the reversal of direct oral anticoagulants in bleeding patients [published online ahead of print 24 September 2018]
.
J Intensive Care Med
.
doi:10.1177/0885066618800657
.
8.
Arachchillage
DRJ
,
Alavian
S
,
Griffin
J
, et al
.
Efficacy and safety of prothrombin complex concentrate in patients treated with rivaroxaban or apixaban compared to warfarin presenting with major bleeding
.
Br J Haematol
.
2019
;
184
(
5
):
808
-
816
.
9.
Berger
K
,
Santibañez
M
,
Lin
L
,
Lesch
CA
.
A low-dose 4F-PCC protocol for DOAC-associated intracranial hemorrhage [published online ahead of print 14 April 2019]
.
J Intensive Care Med
.
doi:10.1177/0885066619840992
.
10.
Dybdahl
D
,
Walliser
G
,
Chance Spalding
M
,
Pershing
M
,
Kincaid
M
.
Four-factor prothrombin complex concentrate for the reversal of factor Xa inhibitors for traumatic intracranial hemorrhage [published online ahead of print 9 January 2019]
.
Am J Emerg Med
.
doi:10.1016/j.ajem.2019.01.008
.
11.
Grandhi
R
,
Newman
WC
,
Zhang
X
, et al
.
Administration of 4-factor prothrombin complex concentrate as an antidote for intracranial bleeding in patients taking direct factor Xa inhibitors
.
World Neurosurg
.
2015
;
84
(
6
):
1956
-
1961
.
12.
Harrison
SK
,
Garrett
JS
,
Kohman
KN
,
Kline
JA
.
Comparison of outcomes in patients with intracranial hemorrhage on factor Xa inhibitors versus vitamin K antagonists treated with 4-factor prothrombin complex concentrate
.
Proc Bayl Univ Med Cent
.
2018
;
31
(
2
):
153
-
156
.
13.
Müller
M
,
Eastline
J
,
Nagler
M
,
Exadaktylos
AK
,
Sauter
TC
.
Application of prothrombin complex concentrate for reversal of direct oral anticoagulants in clinical practice: indications, patient characteristics and clinical outcomes compared to reversal of vitamin K antagonists
.
Scand J Trauma Resusc Emerg Med
.
2019
;
27
(
1
):
48
.
14.
Sheikh-Taha
M
.
Treatment of apixaban- and rivaroxaban-associated major bleeding using 4-factor prothrombin complex concentrate
.
Intern Emerg Med
.
2019
;
14
(
2
):
265
-
269
.
15.
Smith
MN
,
Deloney
L
,
Carter
C
,
Weant
KA
,
Eriksson
EA
.
Safety, efficacy, and cost of four-factor prothrombin complex concentrate (4F-PCC) in patients with factor Xa inhibitor-related bleeding: a retrospective study
.
J Thromb Thrombolysis
.
2019
;
48
(
2
):
250
-
255
.
16.
Schulman
S
,
Kearon
C
;
Subcommittee on Control of Anticoagulation of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis
.
Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients
.
J Thromb Haemost
.
2005
;
3
(
4
):
692
-
694
.
17.
Khorsand
N
,
Majeed
A
,
Sarode
R
,
Beyer-Westendorf
J
,
Schulman
S
,
Meijer
K
;
Subcommittee on Control of Anticoagulation
.
Assessment of effectiveness of major bleeding management: proposed definitions for effective hemostasis: communication from the SSC of the ISTH
.
J Thromb Haemost
.
2016
;
14
(
1
):
211
-
214
.
18.
Sarode
R
,
Milling
TJ
Jr
,
Refaai
MA
, et al
.
Efficacy and safety of a 4-factor prothrombin complex concentrate in patients on vitamin K antagonists presenting with major bleeding: a randomized, plasma-controlled, phase IIIb study
.
Circulation
.
2013
;
128
(
11
):
1234
-
1243
.
19.
Mao
G
,
King
L
,
Young
S
,
Kaplan
R
.
Factor eight inhibitor bypassing agent (FEIBA) for reversal of target-specific oral anticoagulants in life-threatening intracranial bleeding
.
J Emerg Med
.
2017
;
52
(
5
):
731
-
737
.
20.
Connolly
SJ
,
Crowther
M
,
Eikelboom
JW
, et al
;
ANNEXA-4 Investigators
.
Full study report of andexanet alfa for bleeding associated with factor Xa inhibitors
.
N Engl J Med
.
2019
;
380
(
14
):
1326
-
1335
.
21.
Dentali
F
,
Marchesi
C
,
Giorgi Pierfranceschi
M
, et al
.
Safety of prothrombin complex concentrates for rapid anticoagulation reversal of vitamin K antagonists: a meta-analysis
.
Thromb Haemost
.
2011
;
106
(
3
):
429
-
438
.

Competing Interests

Conflict-of-interest disclosure: D.M.S. reports receiving honoraria for participating in consultant meetings from Bayer, BMS-Pfizer, Servier Canada, Leo Pharma, Aspen Pharmaceuticals, Novartis, and Portola Pharmaceuticals. M.K. declares no competing financial interests.

Author notes

Off-label drug use: Prothrombin complex concentrates and activated prothrombin complex concentrate were used for the management of factor Xa inhibitor–associated major bleeding.