Ibrutinib therapy was associated with an increased risk of bleeding in previous trials. In this systematic review and meta-analysis of published trials including patients treated with ibrutinib, the relative risk (95% confidence interval [CI]) of overall bleeding was significantly higher in ibrutinib recipients (2.72 [1.62-6.58]), but major bleeding did not show a significant difference (1.66 [0.96-2.85]). The incidences (95% CI) of major bleeding and any bleeding were 3.0 (2.3-3.7) and 20.8 (19.1-22.1) per 100 patient-years, respectively. This analysis is limited by reporting bias from variable ascertainment of bleeding and lack of allocation concealment in some studies and differing exposures between groups, leading to potential overestimation of event rates in the ibrutinib group.

A 65-year-old man with chronic lymphocytic leukemia (CLL) with 17p deletion on first-line ibrutinib 420 mg daily for 8 weeks presented with a 2-week history of easy bruising and epistaxis. He was previously healthy with no personal or family history of a bleeding disorder and no regular antithrombotic drug use. Laboratory testing revealed a hemoglobin concentration of 8 g/dL, lymphocyte count of 13 × 109 /L, and normal platelet count, international normalized ratio, and activated partial thromboplastin time. What is the role of ibrutinib in this patient’s bleeding symptoms?

Ibrutinib is an orally administered irreversible inhibitor of Bruton tyrosine kinase (BTK). BTK is an important component of the B-cell receptor signaling pathway, and it contributes to the proliferation, survival, migration, and homing of normal and malignant B cells. Ibrutinib has demonstrated substantial clinical benefit in the treatment of several B cell malignancies and has received US Food and Drug Administration approval for treatment naive and relapsed chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL),1,2  relapsed mantle cell lymphoma (MCL),3  and Waldenström macroglobulinemia.4  Additional studies are underway across a range of hematologic malignancies and solid tumors, and the approved indications for ibrutinib therapy are likely to expand in the future.

Early clinical trials of ibrutinib2,5  reported increased rates of major bleeding, including subdural hematomas, gastrointestinal bleeding, and hematuria, leading to the exclusion of patients on a vitamin K antagonist from subsequent studies.6  The true incidence, nature, and risk factors for bleeding among ibrutinib recipients remain poorly characterized owing to heterogeneity in the definitions of major bleeding used in individual studies and the lack of a priori prospective ascertainment of bleeding outcomes.

BTK plays a role in platelet signaling through GP1b and GPVI, which mediate platelet aggregation and adhesion through von Willebrand factor and collagen, respectively. However, the effect of BTK inhibition on platelet function and bleeding risk is not clear. Patients with X-linked agammaglobulinemia do not have increased bleeding despite a congenital absence of functional BTK,7  suggesting that bleeding associated with ibrutinib is more complex than BTK inhibition alone. Although the precise mechanisms for ibrutinib-associated bleeding have not been fully elucidated, published data suggest that both disease- and therapy-related platelet dysfunction may contribute to clinical bleeding. Within a phase 2 trial of single-agent ibrutinib in patients with CLL, Lipsky et al evaluated platelet function and coagulation factors at baseline and 4 weeks after initiation of therapy.8  Reduced platelet function prior to therapy, as measured by prolonged epinephrin closure time, and coagulation activity, as measured by lower factor VIII and von Willebrand factor activity, were associated with a significantly increased risk of bleeding.8  Platelet aggregation was impaired in all patients with CLL compared with healthy controls, and responses to both collagen and adenosine 5′-diphosphate agonists were reduced in ibrutinib-treated and treatment-naive patients compared with normal controls, consistent with a direct disease effect on platelet aggregation. Ibrutinib therapy was associated with further reduction in platelet aggregation with collagen while responses to adenosine 5′-diphosphate improved over time on therapy. Bleeding events appeared to decrease beyond 6 months of ibrutinib therapy, perhaps mediated by improved disease control. Bye et al9  studied the effects of ibrutinib on platelet function by comparing signaling in suspension and during adhesion to immobilized ligands. Defects in GPVI and integrin αIIbβ3 platelet signaling resulted in the formation of unstable thrombi that may contribute to bleeding. These and other findings suggest that deficiencies in several signaling pathways may ultimately contribute to ibrutininb-associated bleeding.

We conducted a systematic review of observational studies and randomized controlled trials (RCTs) with the following objectives: (1) to determine the incidence rate of overall and major bleeding events in clinical trials of ibrutinib and (2) to estimate the risk of overall and major bleeding events among patients treated with ibrutinib compared with alternative treatments. We searched MEDLINE and EMBASE for articles in any language reporting on ibrutinib, Imbruvica, or PCI-32765. Studies were excluded if they were case reports, case series, editorials, or phase 1 and dose-ranging studies or if they did not report bleeding events. Conference abstracts older than 12 months were also excluded, because they were likely to be published in medical journals, and cross-sectional studies were excluded because of an inherent high risk of recall bias. Two reviewers screened each title and abstract independently for eligibility. Disagreements were resolved by a third reviewer. The full texts of eligible papers were obtained and assessed for inclusion in duplicate. In case of overlap between articles reporting on the same cohort, we included the study with the largest cohort. The risk of bias of included studies was assessed using the Cochrane risk of bias tool10  for RCTs and methodology adapted from the American College of Chest Physicians guidelines11  for observational studies. The quality of evidence was then assessed using GRADE (Grading Recommendations Assessment and Development Evidence) criteria.12 

Statistical analysis was performed using STATA 14 (StataCorp, College Station, TX). The agreement between reviewers was measured with the κ statistic. Pooled incidence rates were estimated using the approach of Guevara et al.13  Crude study-specific per-participant bleeding rates were then calculated by dividing the number of incident cases of total bleeding and major bleeding by the total number of person-months follow-up. Incidence rates were pooled after applying the Freeman-Tukey double arcsine transformation to stabilize the variances.14  Bleeding events in comparative studies were pooled (RevMan Version 5.3, The Cochrane Collaboration, 2014) with a fixed effects model, using the Mantel-Haenszel method to estimate the relative risk of bleeding. Heterogeneity was evaluated by Cochran’s Q and the I2 statistic. When heterogeneity was high, an analysis was performed using DerSimonian and Laird random effects models.

The search yielded 1871 unique abstracts. There was close agreement between reviewers for title and abstract screening (κ statistic 0.86). In total, 38 full-text manuscripts and 44 conference abstracts were reviewed. Of these 82 manuscripts, 60 were excluded because they lacked data on bleeding (n = 42), did not study ibrutinib (n = 2), were dose-finding studies (n = 1) or case series (n = 1), or presented data included in another manuscript (n = 14). Twenty-two manuscripts reported on bleeding in 2152 ibrutinib recipients (Table 1). Four were RCTs, also including 759 patients on alternative therapies, 10 were phase 2 studies, 3 were prospective cohorts, and 5 were retrospective cohorts. Sixteen manuscripts were full text, and the other 6 manuscripts were abstracts presented at conferences in the last year. Fifteen studies were in patients with CLL/SLL, 4 in patients with MCL, 2 in patients with Waldenström macroglobulinemia, and 1 in a patient with follicular lymphoma.

Table 1.

Characteristics of included studies

ReferenceDesignDiseaseIndicationAge (y), median (range)Sample sizeFollow-up (mo)Overall bleeding incidence (ibrutinib/control)*Major bleeding incidence (ibrutinib/control)Definition of major bleedingRisk of bias
15 RCT CLL/SLL Relapsed/refractory 67 (30-88) 195 ibrutinib, 196 ofatumumab 9.4 86/24 2/3 Grade ≥3, transfused or hospitalized Uncertain 
RCT CLL/SLL Relapsed/refractory 64 (31-86) 289 ibrutinib, 289 placebo 17 89/42 11/5 Grade ≥3 or CNS Uncertain 
RCT CLL/SLL First line 73 (65-90) 136 ibrutinib, 133 chlorambucil 18.4 — 6/3 Grade ≥3 or CNS Uncertain 
RCT MCL Relapsed/refractory 68 (IQR 13) 139 ibrutinib, 141 temsirolimus 20 — 14/9 Grade ≥3 or CNS High 
16 Phase 2 CLL/SLL Relapsed/refractory 67 (55-84) 24 7.5 13 Grade ≥3 High 
Phase 2 CLL/SLL Both first and relapsed/refractory 65.8 (33-85) 86 24 47 — Unknown High 
17 Phase 2 CLL/SLL Both first and relapsed/refractory 68 (37-84) 132 36 81 10 Unknown High 
26 Phase 2 CLL/SLL Both first and relapsed/refractory 63 (35-82) 40 16.8 — Grade ≥2 High 
29 Phase 2 CLL/SLL Relapsed/refractory 64 144 11.5 — Grade 2/3 High 
28 Phase 2 CLL/SLL Relapsed/refractory N = 31 >65 y 71 12.5 — Grade ≥3 High 
18 Phase 2 Follicular lymphoma First line 58 (32-84) 60 10.2 13 Unknown High 
19 Phase 2 MCL Relapsed/refractory 68 (40-84) 111 26.7 56 Grade ≥3 High 
Phase 2 MCL Relapsed/refractory 67 (45-86) 50 16.5 — Requiring drug discontinuation High 
15 Phase 2 CLL/SLL Relapsed/refractory 66 (37-82) 85 20.9 — Grade ≥3 High 
22 Prospective WM Relapsed/refractory 67 (47-90) 31 7.7 Unknown High 
20 Prospective CLL/SLL Relapsed/refractory 66 (35-83) 88 28 — Unknown High 
Prospective WM Relapsed/refractory 63 (44-86) 63 19.1 — Grade ≥2 High 
24 Retrospective CLL/SLL Both first and relapsed/refractory 66 96 7.6 10 Requiring hospitalization High 
21 Retrospective CLL/SLL Relapsed/refractory Not stated 124 6.4 — Requiring drug discontinuation High 
23 Retrospective CLL/SLL Relapsed/refractory 69 92 6.8 30 — Unknown High 
27 Retrospective CLL/SLL Relapsed/refractory 62 (36-80) 54 9.1 — Requiring drug discontinuation High 
25 Retrospective MCL Relapsed/refractory 69 (35-84) 42 10.7 — Unknown High 
ReferenceDesignDiseaseIndicationAge (y), median (range)Sample sizeFollow-up (mo)Overall bleeding incidence (ibrutinib/control)*Major bleeding incidence (ibrutinib/control)Definition of major bleedingRisk of bias
15 RCT CLL/SLL Relapsed/refractory 67 (30-88) 195 ibrutinib, 196 ofatumumab 9.4 86/24 2/3 Grade ≥3, transfused or hospitalized Uncertain 
RCT CLL/SLL Relapsed/refractory 64 (31-86) 289 ibrutinib, 289 placebo 17 89/42 11/5 Grade ≥3 or CNS Uncertain 
RCT CLL/SLL First line 73 (65-90) 136 ibrutinib, 133 chlorambucil 18.4 — 6/3 Grade ≥3 or CNS Uncertain 
RCT MCL Relapsed/refractory 68 (IQR 13) 139 ibrutinib, 141 temsirolimus 20 — 14/9 Grade ≥3 or CNS High 
16 Phase 2 CLL/SLL Relapsed/refractory 67 (55-84) 24 7.5 13 Grade ≥3 High 
Phase 2 CLL/SLL Both first and relapsed/refractory 65.8 (33-85) 86 24 47 — Unknown High 
17 Phase 2 CLL/SLL Both first and relapsed/refractory 68 (37-84) 132 36 81 10 Unknown High 
26 Phase 2 CLL/SLL Both first and relapsed/refractory 63 (35-82) 40 16.8 — Grade ≥2 High 
29 Phase 2 CLL/SLL Relapsed/refractory 64 144 11.5 — Grade 2/3 High 
28 Phase 2 CLL/SLL Relapsed/refractory N = 31 >65 y 71 12.5 — Grade ≥3 High 
18 Phase 2 Follicular lymphoma First line 58 (32-84) 60 10.2 13 Unknown High 
19 Phase 2 MCL Relapsed/refractory 68 (40-84) 111 26.7 56 Grade ≥3 High 
Phase 2 MCL Relapsed/refractory 67 (45-86) 50 16.5 — Requiring drug discontinuation High 
15 Phase 2 CLL/SLL Relapsed/refractory 66 (37-82) 85 20.9 — Grade ≥3 High 
22 Prospective WM Relapsed/refractory 67 (47-90) 31 7.7 Unknown High 
20 Prospective CLL/SLL Relapsed/refractory 66 (35-83) 88 28 — Unknown High 
Prospective WM Relapsed/refractory 63 (44-86) 63 19.1 — Grade ≥2 High 
24 Retrospective CLL/SLL Both first and relapsed/refractory 66 96 7.6 10 Requiring hospitalization High 
21 Retrospective CLL/SLL Relapsed/refractory Not stated 124 6.4 — Requiring drug discontinuation High 
23 Retrospective CLL/SLL Relapsed/refractory 69 92 6.8 30 — Unknown High 
27 Retrospective CLL/SLL Relapsed/refractory 62 (36-80) 54 9.1 — Requiring drug discontinuation High 
25 Retrospective MCL Relapsed/refractory 69 (35-84) 42 10.7 — Unknown High 

–, Not available; CNS, central nervous system; IQR, interquartile range; WM, Waldenström macroglobulinemia.

*

If only major bleeding is reported, overall bleeding is considered not available.

Open label.

Conference abstracts.

Overall bleeding

Thirteen articles reported on the incidence of overall bleeding6,8,15-25  with a pooled annual incidence of any bleeding of 20.8 per 100 patient-years (95% confidence interval [CI], 19.1-22.1) in ibrutinib-treated patients. Two RCTs6,15  compared the incidence of overall bleeding between ibrutinib and an alternative therapy (Figure 1). The pooled incidence of overall bleeding in patients receiving alternative treatments was 11.6 per 100 patient-years (95% CI, 9.1-14.4). The pooled relative risk of any bleeding with ibrutinib use was 2.72 (95% CI, 1.62-4.58; P = .0002). The I2 statistic for heterogeneity was 75%, and the heterogeneity χ2 was 3.97 (P = .05). The random effects model was used because of the significant heterogeneity of studies.

Figure 1.

Forest plot of relative risk of overall bleeding in RCTs of ibrutinib. M-H, Mantel-Haenszel.

Figure 1.

Forest plot of relative risk of overall bleeding in RCTs of ibrutinib. M-H, Mantel-Haenszel.

Close modal

Major bleeding

Seventeen studies1-6,15-19,22,24,26-29  specifically described major bleeding, with a pooled incidence of 2.76 (95% CI, 2.07-3.53) per 100 patient-years in patients receiving ibrutinib. Major bleeding was classified according to the Common Terminology Criteria for Adverse Events and included the following: (1) grade ≥3 (n = 8),1-3,15,16,28,30,31  (2) grade ≥2 (n = 3),4,26,29  (3) reason for drug discontinuation (n = 2),5,27  and (4) bleeding requiring hospitalization (n = 1).24  In addition, 3 studies did not report a definition of major bleeding; one of these studies17  reported 10 events, including 1 resulting in death and 3 leading to drug discontinuation. In the 4 RCTs reporting on major bleeding,1,3,6,15  the pooled incidence in patients receiving alternative treatments was 1.9 (95% CI, 1.1-2.8) per 100 patient-years. The relative risk of major bleeding in ibrutinib recipients as compared with recipients of an alternative therapy was 1.66 (95% CI, 0.96-2.85; P = .07) using a fixed effects model (Figure 2). The random effects model yielded similar results. The I2 statistic for heterogeneity was 0%, and the heterogeneity χ2 was 1.35 (P = .72).

Figure 2.

Forest plot of relative risk of major bleeding in RCTs of ibrutinib. M-H, Mantel-Haenszel.

Figure 2.

Forest plot of relative risk of major bleeding in RCTs of ibrutinib. M-H, Mantel-Haenszel.

Close modal

This study provides moderate-quality evidence that ibrutinib increases overall bleeding compared with alternative treatments. The observational studies were judged to be at high risk of bias because of the absence of a comparator group. The definitions of major bleeding showed variability among the studies. While most studies used grade ≥3 to define major bleeding events, there was heterogeneity in those definitions and even more in the definition and reporting of minor bleeding. The quality of evidence for RCTs was downgraded due to heterogeneity between studies in reporting of bleeding outcomes. Our meta-analysis was likely underpowered with regards to major bleeding complications, so a difference in the risk of major bleeding between ibrutinib and other treatments cannot be excluded. Some limitations of our analysis should be noted. Bleeding events may have been underestimated in some studies, as they were not captured a priori as study outcomes with clear definitions, systematic data collection, or adjudication processes reported. Lack of concealment of treatment allocation may have led to reporting bias in some trials. Few details were provided regarding factors that may have contributed to bleeding events such as the use of antithrombotic therapy or concurrent severe thrombocytopenia. Finally, in the 4 RCTs, patients were exposed to ibrutinib for longer periods than to the control treatment. This could lead to an overestimation of ibrutinib-related adverse events. Dreyling et al3  performed an exposure-adjusted analysis and revealed bleeding rates of 0.8 per 100 patient-months in the ibrutinib group and 1.1 per 100 patient-months in the temsirolimus group. Exposure-adjusted incidence rates were not reported by the other authors. As a sensitivity analysis, we calculated incidence rates based on the duration of exposure (Table 2). This analysis removes any difference in rates of major bleeding between groups.

Table 2.

Incidence of bleeding adjusted for exposure

ReferenceMedian exposure (mo)Overall bleeding incidence*Major bleeding incidence*
IbrutinibControlIbrutinibControlIbrutinibControl
15 8.4 5.3 63.0 27.6 1.4 3.5 
17.4 7.1 — — 3.0 3.8 
14.4 — — 8.4 25.9 
14.7 12.8 25.1 13.6 3.1 1.7 
ReferenceMedian exposure (mo)Overall bleeding incidence*Major bleeding incidence*
IbrutinibControlIbrutinibControlIbrutinibControl
15 8.4 5.3 63.0 27.6 1.4 3.5 
17.4 7.1 — — 3.0 3.8 
14.4 — — 8.4 25.9 
14.7 12.8 25.1 13.6 3.1 1.7 

–, Not available.

*

Incidence is reported per 100 person-years of exposure.

The incidence rates of overall and major bleeding observed with ibrutinib use are high enough to warrant concern. Although not directly comparable, major bleeding complications in patients receiving anticoagulant therapy for atrial fibrillation occur at rates of 2.1 to 3.6 per 100 patient-years with case fatality rates of 7% to 14% in clinical trials.32-36  With the expected rise in ibrutinib use for the treatment of B-cell lymphoproliferative diseases, bleeding events will likely become an increasingly significant clinical concern, especially among elderly patients requiring concurrent antithrombotic therapy who were underrepresented or excluded from clinical trials.1-5  An increased rate of atrial fibrillation has been observed in clinical trials of ibrutinib.37,38  While there is net benefit from anticoagulation in individuals from the general population with atrial fibrillation and additional stroke risk factors, the increased bleeding risk in ibrutinib-treated patients may alter the risk-benefit ratio of anticoagulant therapy for the prevention of stroke and systemic embolism. To our knowledge, there is no evidence regarding the stroke risk in ibrutinib-treated patients with atrial fibrillation or whether widely used clinical prediction models for stroke, such as CHADS2 or CHADS2Vasc, or bleeding, such as HAS-BLED or ATRIA, are helpful in this setting.39-41  Patients with an indication for warfarin were excluded from the later trials after high rates of bleeding were noticed in earlier studies. Until further data identify the risk factors for bleeding, the decision to initiate anticoagulant therapy should be individualized based on a discussion of patient values and preferences and considering the available evidence regarding the benefits and risks of therapy.

Another relevant clinical scenario is the patient requiring dual antiplatelet therapy (DAPT) after percutaneous coronary intervention with stent placement. Following drug-eluting coronary stent implantation, a minimum of 12 months of DAPT with aspirin and clopidogrel/ticagrelor is preferred for the prevention of late stent thrombosis, which confers a 20% to 45% case-fatality rate. Bare metal stents struts are covered more rapidly by endothelium so are associated with a lower risk of late-stent thrombosis. A minimum of 2 to 4 weeks of DAPT is considered important after bare metal coronary stent implantation, making it a more attractive alternative in the patient with an indication for percutaneous coronary intervention.42 

In the event of clinically relevant bleeding, the ibrutinib product monograph43  advises a dose reduction by 1 tablet, although the effect of this approach on hematologic disease and the risk of subsequent bleeding events are unknown. The indications and dosages of antithrombotic therapies, nonsteroidal anti-inflammatory drugs, and herbal supplements should be reviewed. Drug interactions with inhibitors of CYP3A4 should be explored, as they could increase the concentration of ibrutinib. In cases of major bleeding, supportive therapy should be promptly initiated, and reversible contributing factors addressed. Ibrutinib should be withheld until resolution of the bleeding; the decision to resume ibrutinib will depend on the ongoing bleeding risk and the status of the underlying malignancy. There is a paucity of data regarding the bleeding risk of invasive procedures or surgeries in patients receiving ibrutinib. The product monograph advises temporary interruption of ibrutinib 7 days prior to an invasive procedure.

This review points to the importance of developing standardized reporting of cardiovascular outcomes for oncology studies. Definitions for bleeding events have been developed by the International society of thrombosis and hemostasis44  to be used in studies of products that interfere with coagulation. The use of such definitions would decrease the interstudy variability outcome reporting. Our analysis also revealed that reporting of treatment-emergent adverse events is a source of bias in studies with large difference in exposure time between groups. Intention-to-treat analysis or exposure-adjusted incidences can help to minimize this effect. Bleeding has emerged as an important adverse effect of ibrutinib. Recommendations regarding the approach to bleeding in ibrutinib recipients are largely empiric at present, and more data on the risk of bleeding, bleeding risk factors, and the risks of discontinuing ibrutinib are needed to permit evidence-guided recommendations.

D.P.L. is supported by the E. J. Moran Campbell Career Award from McMaster University and by the Hamilton Health Sciences RFA Strategic Initiative Program. C.H. is supported by a fellowship from the Department of Oncology, McMaster University. D.S. is supported by a fellowship award from the Canadian Institutes of Health Research.

Contribution: F.C., D.P.L., and D.S. contributed to data extraction; F.C. and D.P.L. analyzed the data; D.S. and F.C. assessed the risk of bias; and all authors contributed to writing the paper.

Conflict-of-interest disclosure: D.P.L. has received honoraria from Janssen Pharmaceuticals. G.F. has received honoraria and research funding from Janssen Pharmaceuticals. The remaining authors declare no competing financial interests.

Correspondence: François Caron, David Braley Building, Hamilton General Hospital, 237 Barton St E, Hamilton, ON L8L 2X2, Canada; e-mail: francois.caron@phri.ca.

1.
Burger
JA
,
Tedeschi
A
,
Barr
PM
, et al
;
RESONATE-2 Investigators
.
Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia
.
N Engl J Med
.
2015
;
373
(
25
):
2425
-
2437
.
2.
Byrd
JC
,
Furman
RR
,
Coutre
SE
, et al
.
Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia
.
N Engl J Med
.
2013
;
369
(
1
):
32
-
42
.
3.
Dreyling
M
,
Jurczak
W
,
Jerkeman
M
, et al
.
Ibrutinib versus temsirolimus in patients with relapsed or refractory mantle-cell lymphoma: an international, randomised, open-label, phase 3 study. [Erratum appears in Lancet. 2016;387(10020):750.]
Lancet
.
2016
;
387
(
10020
):
770
-
778
.
4.
Treon
SP
,
Tripsas
CK
,
Meid
K
, et al
.
Ibrutinib in previously treated Waldenström’s macroglobulinemia
.
N Engl J Med
.
2015
;
372
(
15
):
1430
-
1440
.
5.
Wang
ML
,
Lee
H
,
Chuang
H
, et al
.
Ibrutinib in combination with rituximab in relapsed or refractory mantle cell lymphoma: a single-centre, open-label, phase 2 trial
.
Lancet Oncol
.
2016
;
17
(
1
):
48
-
56
.
6.
Chanan-Khan
A
,
Cramer
P
,
Demirkan
F
, et al
;
HELIOS investigators
.
Ibrutinib combined with bendamustine and rituximab compared with placebo, bendamustine, and rituximab for previously treated chronic lymphocytic leukaemia or small lymphocytic lymphoma (HELIOS): a randomised, double-blind, phase 3 study
.
Lancet Oncol
.
2016
;
17
(
2
):
200
-
211
.
7.
Oda
A
,
Ikeda
Y
,
Ochs
HD
, et al
.
Rapid tyrosine phosphorylation and activation of Bruton’s tyrosine/Tec kinases in platelets induced by collagen binding or CD32 cross-linking
.
Blood
.
2000
;
95
(
5
):
1663
-
1670
.
8.
Lipsky
AH
,
Farooqui
MZ
,
Tian
X
, et al
.
Incidence and risk factors of bleeding-related adverse events in patients with chronic lymphocytic leukemia treated with ibrutinib
.
Haematologica
.
2015
;
100
(
12
):
1571
-
1578
.
9.
Bye
AP
,
Unsworth
AJ
,
Vaiyapuri
S
,
Stainer
AR
,
Fry
MJ
,
Gibbins
JM
.
Ibrutinib Inhibits Platelet Integrin αIIbβ3 Outside-In Signaling and Thrombus Stability But Not Adhesion to Collagen
.
Arterioscler Thromb Vasc Biol
.
2015
;
35
(
11
):
2326
-
2335
.
10.
Higgins
JP
,
Altman
DG
,
Gøtzsche
PC
, et al
;
Cochrane Bias Methods Group
;
Cochrane Statistical Methods Group
.
The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials
.
BMJ
.
2011
;
343
:
d5928
.
11.
Guyatt
GH
,
Norris
SL
,
Schulman
S
, et al
.
Methodology for the development of antithrombotic therapy and prevention of thrombosis guidelines: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines
.
Chest
.
2012
;
141
(
2 suppl
):
53S
-
70S
.
12.
Balshem
H
,
Helfand
M
,
Schünemann
HJ
, et al
.
GRADE guidelines: 3. Rating the quality of evidence
.
J Clin Epidemiol
.
2011
;
64
(
4
):
401
-
406
.
13.
Guevara
JP
,
Berlin
JA
,
Wolf
FM
.
Meta-analytic methods for pooling rates when follow-up duration varies: a case study
.
BMC Med Res Methodol
.
2004
;
4
:
17
.
14.
Freeman
M
,
Tukey
J
.
Transformations related to the angular and the square root
.
Ann Math Stat
.
1950
;
21
:
607
-
611
.
15.
Byrd
JC
,
Brown
JR
,
O’Brien
S
, et al
;
RESONATE Investigators
.
Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia
.
N Engl J Med
.
2014
;
371
(
3
):
213
-
223
.
16.
Kazianka
L
,
Drucker
C
,
Skrabs
C
, et al
.
Ristocetin-induced platelet aggregation for monitoring of bleeding tendency in ibrutinib-treated patients with chronic lymphocytic leukemia [abstract]
.
Blood
.
2015
;
126
(
23
).
Abstract 718
.
17.
Byrd
JC
,
Furman
RR
,
Coutre
SE
, et al
.
Three-year follow-up of treatment-naïve and previously treated patients with CLL and SLL receiving single-agent ibrutinib
.
Blood
.
2015
;
125
(
16
):
2497
-
2506
.
18.
Fowler
N
,
Nastoupil
L
,
De Vos
S
, et al
.
Ibrutinib plus rituximab in treatment-naive patients with follicular lymphoma: results from a multicenter, phase 2 study [abstract]
.
Blood
.
2015
;
126
(
23
).
Abstract 470
.
19.
Wang
ML
,
Blum
KA
,
Martin
P
, et al
.
Long-term follow-up of MCL patients treated with single-agent ibrutinib: updated safety and efficacy results
.
Blood
.
2015
;
126
(
6
):
739
-
745
.
20.
Thompson
PA
,
O’Brien
SM
,
Wierda
WG
, et al
.
Complex karyotype is a stronger predictor than del(17p) for an inferior outcome in relapsed or refractory chronic lymphocytic leukemia patients treated with ibrutinib-based regimens
.
Cancer
.
2015
;
121
(
20
):
3612
-
3621
.
21.
Parikh
SA
,
Chaffee
KR
,
Call
TG
, et al
.
Ibrutinib therapy for chronic lymphocytic leukemia (CLL): An analysis of a large cohort of patients treated in routine clinical practice [abstract]
.
Blood
.
2015
;
126
(
23
).
Abstract 2935
.
22.
Dimopoulos
MA
,
Trotman
J
,
Tedeschi
A
, et al
.
Ibrutinib therapy in rituximab-refractory patients with waldenstrom’s macroglobulinemia: initial results from an international, multicenter, open-label phase 3 substudy (innovatetm)
.
Blood
.
2015
;
126
:
2745
.
23.
Hansson
L
,
Winqvist
M
,
Asklid
A
, et al
.
Real-world results on ibrutinib in patients with relapsed or refractory chronic lymphocytic leukemia (CLL): data from 97 Swedish patients treated in a compassionate use program [abstract]
.
Blood
.
2015
;
126
(
23
).
Abstract 1745
.
24.
Finnes
HD
,
Chaffee
KG
,
Call
TG
, et al
.
The importance of pharmacovigilance during ibrutinib therapy for chronic lymphocytic leukemia (CLL) in routine clinical practice [abstract]
.
Blood
.
2015
;
126
(
23
).
Abstract 717
.
25.
Cheah
CY
,
Chihara
D
,
Romaguera
JE
, et al
.
Patients with mantle cell lymphoma failing ibrutinib are unlikely to respond to salvage chemotherapy and have poor outcomes
.
Ann Oncol
.
2015
;
26
(
6
):
1175
-
1179
.
26.
Burger
JA
,
Keating
MJ
,
Wierda
WG
, et al
.
Safety and activity of ibrutinib plus rituximab for patients with high-risk chronic lymphocytic leukaemia: a single-arm, phase 2 study
.
Lancet Oncol
.
2014
;
15
(
10
):
1090
-
1099
.
27.
Sandoval-Sus
JD
,
Chavez
JC
,
Dalia
S
, et al
.
Outcomes of patients with relapsed/refractory chronic lymphocytic leukemia after ibrutinib discontinuation outside clinical trials: A single institution experience [abstract]
.
Blood
.
2015
;
126
(
23
).
Abstract 2945
.
28.
Jaglowski
SM
,
Jones
JA
,
Nagar
V
, et al
.
Safety and activity of BTK inhibitor ibrutinib combined with ofatumumab in chronic lymphocytic leukemia: a phase 1b/2 study
.
Blood
.
2015
;
126
(
7
):
842
-
850
.
29.
Stilgenbauer
S
,
Jones
JA
,
Coutre
SE
, et al
.
Results from the phase 2 RESONATETM-17 trial: efficacy and safety of Ibrutinib in patients with relapsed or refractory chronic lymphocytic leukemia or small lymphocytic lymphoma with Del17p [abstract]
.
Oncol Res Treat
.
2015
;
38
:
92
.
30.
Chanan-Khan
A
,
Demirkan
PCF
,
Fraser
G
, et al
.
Insights into the management of adverse events in patients with previously treated chronic lymphocytic leukemia/small lymphocytic lymphoma: experience from the phase 3 helios study of ibrutinib combined with bendamustine/rituximab
.
Blood
.
2015
;
126
:
2936
.
31.
Wang
M
,
Chuang
HLH
,
Wagner-Bartak
N
, et al
.
Ibrutinib in combination with rituximab for relapsed mantle cell lymphoma: an update for a phase II clinical trial
.
Clin Lymphoma Myeloma Leuk
.
2015
;
15
(
suppl 2
):
S65
.
32.
Majeed
A
,
Hwang
HG
,
Connolly
SJ
, et al
.
Management and outcomes of major bleeding during treatment with dabigatran or warfarin
.
Circulation
.
2013
;
128
(
21
):
2325
-
2332
.
33.
Giugliano
RP
,
Ruff
CT
,
Braunwald
E
, et al
;
ENGAGE AF-TIMI 48 Investigators
.
Edoxaban versus warfarin in patients with atrial fibrillation
.
N Engl J Med
.
2013
;
369
(
22
):
2093
-
2104
.
34.
Connolly
SJ
,
Ezekowitz
MD
,
Yusuf
S
, et al
;
RE-LY Steering Committee and Investigators
.
Dabigatran versus warfarin in patients with atrial fibrillation
.
N Engl J Med
.
2009
;
361
(
12
):
1139
-
1151
.
35.
Granger
CB
,
Alexander
JH
,
McMurray
JJ
, et al
;
ARISTOTLE Committees and Investigators
.
Apixaban versus warfarin in patients with atrial fibrillation
.
N Engl J Med
.
2011
;
365
(
11
):
981
-
992
.
36.
Patel
MR
,
Mahaffey
KW
,
Garg
J
, et al
;
ROCKET AF Investigators
.
Rivaroxaban versus warfarin in nonvalvular atrial fibrillation
.
N Engl J Med
.
2011
;
365
(
10
):
883
-
891
.
37.
Farooqui
M
,
Valdez
J
,
Soto
S
,
Bray
A
,
Tian
X
,
Wiestner
A
.
Atrial fibrillation in CLL/SLL patients on ibrutinib
.
Blood
.
2015
;
126
(
23
):
2933
.
38.
Leong
DP
,
Caron
F
,
Hillis
C
, et al
.
The risk of atrial fibrillation with ibrutinib use: a systematic review and meta-analysis
.
Blood
.
2016
;
128
(
1
):
138
-
140
.
39.
Pisters
R
,
Lane
DA
,
Nieuwlaat
R
,
de Vos
CB
,
Crijns
HJ
,
Lip
GY
.
A novel user-friendly score (HAS-BLED) to assess 1-year risk of major bleeding in patients with atrial fibrillation: the Euro Heart Survey
.
Chest
.
2010
;
138
(
5
):
1093
-
1100
.
40.
Fang
MC
,
Go
AS
,
Chang
Y
, et al
.
A new risk scheme to predict warfarin-associated hemorrhage: the ATRIA (Anticoagulation and Risk Factors in Atrial Fibrillation) study
.
J Am Coll Cardiol
.
2011
;
58
(
4
):
395
-
401
.
41.
Naccarelli
GV
,
Panaccio
MP
,
Cummins
G
,
Tu
N
.
CHADS2 and CHA2DS2-VASc risk factors to predict first cardiovascular hospitalization among atrial fibrillation/atrial flutter patients
.
Am J Cardiol
.
2012
;
109
(
10
):
1526
-
1533
.
42.
Fihn
SD
,
Gardin
JM
,
Abrams
J
, et al
;
American College of Cardiology Foundation/American Heart Association Task Force
.
2012 ACCF/AHA/ACP/AATS/PCNA/SCAI/STS guideline for the diagnosis and management of patients with stable ischemic heart disease: a report of the American College of Cardiology Foundation/American Heart Association task force on practice guidelines, and the American College of Physicians, American Association for Thoracic Surgery, Preventive Cardiovascular Nurses Association, Society for Cardiovascular Angiography and Interventions, and Society of Thoracic Surgeons
.
Circulation
.
2012
;
126
(
25
):
e354
-
e471
.
43.
Janssen
.
Imbruvica product monograph
. .
44.
Schulman
S
,
Angerås
U
,
Bergqvist
D
,
Eriksson
B
,
Lassen
MR
,
Fisher
W
;
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 surgical patients
.
J Thromb Haemost
.
2010
;
8
(
1
):
202
-
204
.

Author notes

*

F.C. and D.P.L. contributed equally to this study.