You are on page 1of 12

Leukemia & Lymphoma

ISSN: 1042-8194 (Print) 1029-2403 (Online) Journal homepage: http://www.tandfonline.com/loi/ilal20

Cardiac side effects of bruton tyrosine kinase (BTK)


inhibitors

Chloe Pek Sang Tang, Julie McMullen & Constantine Tam

To cite this article: Chloe Pek Sang Tang, Julie McMullen & Constantine Tam (2017):
Cardiac side effects of bruton tyrosine kinase (BTK) inhibitors, Leukemia & Lymphoma, DOI:
10.1080/10428194.2017.1375110

To link to this article: http://dx.doi.org/10.1080/10428194.2017.1375110

View supplementary material

Published online: 13 Sep 2017.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ilal20

Download by: [Imperial College London Library] Date: 14 September 2017, At: 04:49
LEUKEMIA & LYMPHOMA, 2017
https://doi.org/10.1080/10428194.2017.1375110

REVIEW

Cardiac side effects of bruton tyrosine kinase (BTK) inhibitors


Chloe Pek Sang Tanga, Julie McMullenb and Constantine Tama
a
Department of Haematology, St Vincent’s Hospital Melbourne, Fitzroy, Australia; bBaker IDI Heart and Diabetes Institute, Melbourne,
Australia

ABSTRACT ARTICLE HISTORY


The development of bruton tyrosine kinase inhibitors (BTKi) has been a significant advancement Received 18 July 2017
in the treatment of chronic lymphocytic leukemia and related B-cell malignancies. As experience Revised 24 August 2017
in using ibrutinib increased, the first drug to be licensed in its class, atrial fibrillation (AF) Accepted 30 August 2017
emerged as an important side effect. The intersection between BTKi therapy for B-cell malignan-
KEYWORDS
Downloaded by [Imperial College London Library] at 04:49 14 September 2017

cies and AF represents a complex area of management with scant evidence for guidance. Bruton kinase inhibitor;
Consideration needs to be taken regarding the interplay of increased bleeding risk versus atrial fibrillation; ibrutinib
thromboembolic complications of AF, drug interactions between ibrutinib and anticoagulants
and antiarrhythmic agents, and the potential for other, as yet seldom reported cardiac side
effects. This review describes the current knowledge regarding BTKi and potential pathophysio-
logic mechanisms of AF and discusses the management of BTKi-associated AF. Finally, a review
of the second generation BTKi is provided and gaps in knowledge in this evolving field are
highlighted.

Background microenvironment are pivotal in the pathogenesis of


the disease [14]. Autonomous BCR-signaling pathways
Mechanism of action of ibrutinib
promote proliferation and survival of CLL cells [15].
Ibrutinib is an orally bioavailable bruton kinase inhibi- The BCR is a multimer complex of membrane immuno-
tor (BTKi), which forms an irreversible covalent bond globulin bound to heterodimers CD79a/CD79b.
to BTK at the Cysteine-481 residue [1,2]. However, its Antigen ligation triggers the formation of BCR signalo-
binding profile is not restricted to BTK as it inhibits some, consisting of protein tyrosine kinase (LYN),
other kinases such as ITK (interleukin-2-inducible T-cell spleen tyrosine kinase (SYK), BTK, phospholipase Cc2
kinase), tec protein tyrosine kinase (TEC), BMX and epi- (PLCc2) and phosphoinositide 3-kinase (PI3K)
dermal growth factor receptor (EGFR) [3]. It has proven (Figure 1). Phosphorylation of the signalosome results
efficacy in several B-cell malignancies, including in further activation of two main pathways: BTK and
chronic lymphocytic leukemia (CLL), mantle cell lymph- PI3K-AKT [14].
oma (MCL), Waldenstrom’s macroglobulemia and mar- BTK is a nonreceptor tyrosine kinase in the Tec fam-
ginal zone lymphoma, both in the upfront setting and ily of tyrosine kinases. BTK is primarily expressed in B
relapsed or refractory (R/R) setting [4–8]. Ibrutinib lymphocytes but not T lymphocytes and plasma cells
induces apoptosis and reduces proliferation of malig- [16]. Following activation of BTK by SYK and LYN, BTK
nant CLL cells by downregulating B-cell receptor (BCR) in turn phosphorylates PLCc2 via B-cell linker protein
signaling and thwarts the supportive tissue microenvir- (BLNK). This subsequently leads to downstream
onment for malignant cells by targeting chemokine- initiation of a signaling cascade consisting of nuclear
controlled adhesion and migration of malignant cells factor-jB (NF-jB), mitogen-activated protein kinase
[9–11]. (MAPK) and protein kinase Cb (PK Cb), resulting in cell
CLL is the most common form of adult leukemia proliferation and transcriptional activation (Figure 1)
[12]. It is characterized by accumulation of malignant [17,18].
mature B cells in the bone marrow, peripheral blood, PI3Ks are divided into three classes (I, II and III) and
lymph node and spleen [13]. BCR-signaling pathways class I is further comprised of four isoforms (a, b, c, d).
and the interactions of the leukemic cells with its The predominant form expressed by hematopoietic

CONTACT Constantine Tam Constantine.Tam@petermac.org 41 Victoria Parade, Fitzroy 3065, Victoria, Australia
ß 2017 Informa UK Limited, trading as Taylor & Francis Group
2 C. P. S. TANG ET AL.
Downloaded by [Imperial College London Library] at 04:49 14 September 2017

Figure 1. B-cell receptor signalling pathways in chronic lymphocytic leukaemia. Abbreviations: BMSC: bone marrow stromal cells;
NLC: nurselike cell; FDC: follicular dendritic cell; NK cell: natural killer cell.

cells is the PI3Kd. PI3K forms PIP3 which in turns acti- Ibrutinib and CLL
vates the Akt pathway by recruiting Akt to the plasma
Ibrutinib was first granted accelerated approval by the
membrane, thereby inhibiting apoptosis and promot-
US Food and Drug Administration (FDA) following the
ing cell survival by phosphorylating the proapoptotic
pivotal trial published by Byrd et al. [4,22]. A total of
proteins Bad and FOX03a [19].
101 relapsed or refractory (R/R) patients received ibru-
Both BTK and PI3K pathways regulate CLL cell
interactions with its tissue microenvironment [11,20]. tinib, with the majority of them being high-risk
CLL cells recirculate between peripheral blood, lymph patients (34% del (17p); 78% unmutated immuno-
nodes and bone marrow within a tumor microenvir- globulin heavy chain (IGHV)). The overall response rate
onment. This process of tissue homing and CLL traf- (ORR) was 90% and estimated progression-free survival
ficking is a tightly regulated process whereby CLL (PFS) at 30 months was 69%. For patients with del
cells engage in crosstalk with a host of accessory (17p) and del (11q), the median PFS was 28 months
cells (e.g. nurse-like cells, T lymphocytes and bone and 38.7 months, respectively [4,22]. Subsequently,
marrow stromal cells) in these proliferating compart- several trials have validated the efficacy of ibrutinib
ments. BTK and PI3K integrate signals from cell sur- both in the upfront setting and in the R/R setting,
face receptors including CXCR4/5, CD40 and integrin, either as a single agent or as combination therapy
enhancing chemostaxis to CXCL 12/13 expressed by (Table 1). These studies heralded a paradigm shift for
the accessory cells. These interactions serve to the treatment of CLL patients, in particular those with
improve CLL longevity and bypass immune-mediated high-risk features such as TP53 dysregulation (deletion
destruction [9,10,21]. or mutation), unmutated IGHV, those who are
CARDIOTOXICITY OF BTK 3

conventionally refractory to fludarabine-based chemo-

97% and 79% for R/R

CR: complete response; mths: months; ORR: overall response rate; OS: overall survival; PFS: progression-free survival; Pts: patients; RCT: randomized controlled trial; R/R: relapsed or refractory; TN: treatment naïve;
At 30mo, OS for TN was
therapy, as well as older or frail patients who may not

92.3 vs. 85 vs. 87.5


Not reached vs. 8.1 90 vs. 79 at 12 mo
84 vs 74 at 24 mo
tolerate toxicity related to standard chemotherapy
Not reached in

75% at 24 mo

83.8 at 18 mo
OS

both arms
[30]. High-risk patients treated with first-line chemo-

at 12 mo
95 vs. 84
therapy generally experienced abysmal outcomes with
reported median progression free survival for del 17p
and TP53 mutation of 19.2 months and 23.3 months

91 vs 80 at 24 mo
respectively [31]. Similarly, median overall survival was

75 at 12 mo
not reached

88.7 vs. 85 vs.


63% at 24 mo
23% for TN and 96% at 30 mo

78% at 18 mo
poor for del (17p) and TP53 mutation patients, at 19.2
PFS

months and 30.2 months, respectively [31]. The


89 vs. 84
13.3 vs.

updated result of the CLL8 trial suggests that FCR (flu-


darabine, cyclophosphamide and rituximab) chemoim-
munotherapy may represent the optimal first-line
0 vs. 5 vs. 0
2.8 vs. 10.4

7% for R/R

treatment for young and fit patients without adverse


8 vs 2
18 vs
8%
CR

8 high-risk features, especially in the era whereby min-


Downloaded by [Imperial College London Library] at 04:49 14 September 2017

imal residual disease (MRD) negativity may signify a


curative possibility [32,33]. However, for the older or
100 vs. 84 vs. 75
67.8 vs. 82.7

frail patients, the high morbidity from infections (13%)


92 vs. 36

97 vs. 80

63 vs. 4
83%
89%
ORR

and prolonged neutropenia (4%) of fludarabine-based


95

chemotherapy may prove to be intolerable [32].


Most of the longer-term outcomes from recent trials
as illustrated in Table 1 suggest that ibrutinib is very
32 vs. 33
Del 17p
0

100%
27

100%

44

40

well tolerated with a rapid and durable response. The


most common side effects from ibrutinib treatment
Follow-up

are diarrhea (59%), upper respiratory tract infection


Table 1. Trials demonstrating efficacy of ibrutinib in both upfront and relapsed or refractory CLL.

(mths)

28.6

12.5

16.8
9.4
17

28
36

24

(47%), bruising (51%) and fatigue (33%) [34]. These


events were predominantly mild (grade 1–2) in nature.
(yrs; median)

AF and bleeding are emerging as notable grade 3 side


Age

effects that warrant vigilant monitoring and AF will be


64

64
68

73

62

67
64

63

the focus of this review.


Bendamustine þ rituximab (BR)þ

Ibrutinib and mantle cell lymphoma (MCL)


placebo vs BR þ Ibrutinib

In 2013, ibrutinib first gained accelerated approval


Comparator

from FDA following pilot data published by Wang


et al. In this phase-2 study, 111 patients with relapsed
Ofatumumab
Chlorambucil

or refractory mantle cell lymphoma received a daily


dose of 560 mg and were followed up for a median
None
None

None

None

None

duration of 15.3 months. An overall response rate of


60% was achieved of which 21% achieved complete
ibrutinib and rituximab,
40 Phase-2, single-arm with
145 Phase-2, single-arm, R/R
132 Phase-2, single-arm, TN

85 Phase-2, single-arm, TN

remission. The estimated progression-free survival was


71 Phase-1b/2, single-arm,
R/R ibrutinib þ of
Study Design

13.9 months and overall survival was 58% at 18


months [35]. More recently, the updated data from
R/R and TN
atumumab

this cohort of patients with median follow-up of 26.7


and R/R

and R/R
578 RCT, R/R

391 RCT, R/R


269 RCT, TN

months showed a durable response with an overall


response rate of 67%, 24-month progression-free
survival and overall survival rate of 31% and 47%,
No.
pts

respectively [6].
Jaglowski 2015 [28]
Farooqui 2015 [26]

The efficacy of ibrutinib in treating R/R advanced


O’Brien 2016 [24]

Burger 2014 [29]


Byrd 2014 [27]

MCL was further validated by a phase-3, randomized,


Byrd 2015 [4]
Chanan-Khan
2016 [23]

2016 [25]

open-label trial comparing ibrutinib with temsirolimus.


Barr et al.,

yrs: years.

The ibrutinib arm demonstrated superior progression-


Trial

free survival (14.6 months vs. 6.2 months) and less


4 C. P. S. TANG ET AL.

Table 2. Rate of detection of AF in ibrutinib trials.


Trial No of patients Study design Median follow-up Rate of AF AF grade 3 Discontinuation rate from AF
Coutre 2017 [45] 94 Phase–1b/2, Single arm 44mo 6% 6%
Chanan-Khan 2016 [23] 578 RCT 17 mo 7% Not reported 1%
O’Brien 2016 [24] 144 Phase-2, single arm 27.6mo 7% 6% 7% had dose reduction
Jain 2016 [46] 40 Phase-2, single arm (IR) 47 mo 7% 2%
Barr 2015 [25] 269 RCT 28.5 10% 6%
Farooqui 2015 [26] 85 Phase-2, single arm 24mo 2% 0%
Byrd 2015 [4] 132 Phase-2, single arm 36mo 6% 6%
Jaglowski [28] 71 Phase-1b/2, single arm 12.5mo 8.5% 3%
Byrd 2014 [27] 391 RCT 9.4mo 5% 3%
AF: atrial fibrillation; mo: months; RCT: randomized controlled trial.

grade 3 or higher adverse events (68% vs. 87%) after a Table 3. Rate of AF in phase-3 trials.
median follow-up of 20 months [36]. Trial Ibrutinib arm (%) Comparator arm (%)
Byrd 2014 (RESONATE) [27] 3 0
Downloaded by [Imperial College London Library] at 04:49 14 September 2017

Burger 2015 (RESONATE-2) [5] 6 1


Ibrutinib and Waldenstrom’s macroglobulinemia Chanan-Khan 2016 (HELIOS) [23] 7 2
Dreyling 2015 [36] 3 1
(WM)
Waldenstrom’s macroglobulinemia (WM) is an IgM- ibrutinib-induced AF [41]. In a recent meta-analysis of
secreting lymphoplasmacytic lymphoma (LPL) [37]. The 20 studies investigating the impact of AF in ibrutinib-
L265P mutation in MYD88 is the most common acti- treated patients, the pooled rate of AF was 3.3 per
vating somatic mutation in WM, present in more than 100 person-years over a median follow-up of 26
90% of patients [38,39]. Ibrutinib induces apoptosis of months. This was comparatively higher than the noni-
WM cells by inhibiting the activation of nuclear factor- brutinib-treated patients and age-matched general
jB (NF-jB) via the bruton tyrosine kinase’s pathway. population (0.84 per 100 person-years and 0.55 per
Following the landmark study published by Treon 100 person-years, respectively) [42]. Similarly, Shanafelt
et al., ibrutinib was approved for use in WM patients. et al. reported 10-year retrospective data on the preva-
Significant improvement in overall response rate lence of AF in 2292 newly diagnosed patients on ibru-
occurred in all genomic subgroups. In particular, tinib without prior history of AF and found an
responses occurred more rapidly in patients with a incidence rate of 6.1% [43]. Another study reported
MYD88L265P mutation and wild-type CXCR4, as CXCR4 figures as high as 16% after a median follow-up of 28
mutation may confer resistance to ibrutinib. In CXCR4 months [44]. Data from long-term follow-up studies
wild-type patients, the ORR was 100%. Comparatively,
suggest that most of these AF events are of grade 3
patients with both MYD88L265P and CXCR4 mutations’
and would have warranted either treatment reduction
had lower ORR of 85.7%. The two-year progression-
or cessation (Table 2). All major phase–3 trials
free survival and overall survival among all patients
evidently showed that AF rates were unequivocally
were 69.1% and 95.2%, respectively [7]. A further study
elevated compared to the control arms (Table 3). The
of patients with rituximab-refractory WM confirmed
true prevalence of AF may be underrepresented in
the high single-agent activity of ibrutinib in WM [40].
these trials, as systematic AF surveillance was not
conducted.
Ibrutinib and AF
Evidence from trials Implications of AF
Ibrutinib has proven to be highly efficacious with The implications of the link between ibrutinib and AF
remarkable depth of response in B-cell malignancies are multifold. Firstly, the most common reason for
with an acceptable toxicity profile. However, AF ibrutinib discontinuation was treatment-related
appears to be a significant treatment-emergent side toxicity, with AF being the most common cause of
effect that warrants closer inspection. Interestingly, toxicity-related treatment discontinuation [47].
this phenomena is not seen in patients with X-linked Discontinuation rate has been reported as high as 32%
agammaglobulinemia (XLA), a primary immunodefi- from several pivotal studies [46,48,49]. Although the
ciency disease characterized by BTK deficiency second- outcomes of patients who discontinued treatment due
ary to BTK gene mutations, implying that other to treatment intolerance appeared to be superior com-
kinases may be involved in the pathophysiology of pared to those due to Richter transformation (RT) or
CARDIOTOXICITY OF BTK 5

Table 4. Rate of major bleeding from major ibrutinib trials. was 61 years and the median time from initiation of
Major bleeding ibrutinib to these cardiac events was 65 days. Thirteen
Trial Study design (grade 3) rate
of 17 of these patients had no previous cardiac history.
Chanan-Khan (HELIOS) [88] RCT 4%
Byrd et al. 2015 [4] Phase-2, single arm 8% Extensive cardiac workup in patients who developed
Barr (RESONATE 2) [90] RCT 6% ventricular arrhythmia revealed no other provoking
Byrd (RESONATE) [62] RCT 1%
Wang et al. 2015 [6] Phase-2, single arm 6%
factor apart from ibrutinib in 10 out of 11 patients.
Dreyling 2016 [29] RCT 10% Five patients stopped their ibrutinib permanently and
two patients resumed their ibrutinib but had recur-
rence of ventricular arrhythmia despite antiarrhythmic
disease progression, they still collectively performed
agents. In a pooled data of published clinical trials, the
worse than patients who did not have treatment inter-
incidence rates of sudden cardiac death was 788
ruption or cessation [50,51]. In a retrospective study
events per 100,000 person-years for patients on ibruti-
highlighting the impact of AF on patients receiving
nib compared to 200–400 events per 100,000 person-
ibrutinib, patients whose treatment was interrupted
years for the age-matched general population [55].
had inferior progression-free survival compared to
With increasing use of ibrutinib in the future, we will
Downloaded by [Imperial College London Library] at 04:49 14 September 2017

those who proceeded with full dose or dose reduction


likely encounter more of these cardiac events. The
(median 19 months vs. 27 months) [52]. Jain et al ana-
exact pathophysiology of ventricular arrhythmia and
lyzed survival outcomes for 320 patients after ibrutinib
sudden cardiac death is unknown. It will be interesting
discontinuation based on the cause of discontinuation.
to explore if AF and ventricular arrhythmias secondary
The median time to discontinuation was 15 months
to ibrutinib arise from a common mechanism. The true
and after a median follow-up of 38 months, the
prevalence of these events will need to be determined
median survival was 33 months for ibrutinib intoler-
in prospective trials utilizing systematic cardiac
ance, 16 months for disease progression and 2 months
surveillance.
for RT [48].
Secondly, AF is associated with increased stroke
risk, cardiomyopathy and mortality [52,53]. The median Pathophysiology of AF
time from initiation of treatment to onset of AF was In light of the magnitude and impact of AF and poten-
3.8 months and in the majority of events, medical tial for cardiomyopathy for patients treated with ibruti-
therapy failed to prevent recurrence of AF [52]. This nib, it is imperative that the exact pathophysiology by
contrasts with Byrd’s reporting of no detectable events which ibrutinib contributes to cardiac side effects be
of AF after 2 years of treatment, although this may elucidated. McMullen et al. hypothesized that ibrutinib
just reflect the high dropout rate (only 17% of patients causes AF by inhibiting the PI3K–Akt pathway in the
remained on treatment after 3 years) [4]. Shanafelt heart [57]. There is evidence from other cell types of
et al. identified older age, male sex, valvular heart dis- crosstalk between BTK and TEC and the PI3K–Akt path-
ease and hypertension as risk factors for the develop- way [58]. Reduced PI3K signaling in the heart can lead
ment of AF in this unique group [43]. Very recently, to an increased susceptibility to AF [59]. BTK and TEC
baseline (preibrutinib) left atrial abnormality on elec- transcripts are present in human heart tissue, and
trocardiogram (i.e. ‘p mitrale’), prolonged PR interval increased expression was identified in patients with
and lead V1-biphasic P wave were found in a retro- AF, which may signify a cardioprotective role in the
spective case–control study to be associated with a heart in preventing both AF and stress-induced cardio-
6.6-fold increase in the risk of AF arising during ibruti- myopathy [57]. Ibrutinib was found to inhibit PI3K–Akt
nib therapy [54]. This hypothesis generating study signaling in isolated rat neonatal cardiac myocytes
underscores the need for closer evaluation of baseline [57]. In another study, ibrutinib prolonged and trig-
cardiac investigations in order to develop predictive gered abnormal action potentials in isolated adult
models to identify high AF risk patients. mouse and rabbit cardiac myocytes. These defects
More concerningly, there have been reports of sud- were reversed by increasing PI3K by adding PIP3 to
den cardiac deaths and ventricular arrhythmias associ- the patch pipette [60]. The role of PI3K in ibrutinib-
ated with ibrutinib use [55,56]. Retrospective review of induced AF is further reinforced by the fact that idela-
FDA Adverse Event Reporting System (FAERS) from lisib, being a PI3K inhibitor (inhibiting PI3Kd as
2013 to 2015 identified a total of 11 cases of ventricu- opposed to ibrutinib, which affects PI3Ka), does not
lar arrhythmias including ventricular tachycardia (VT) cause cardiotoxicity [61,62]. Clearly, further in-depth
and ventricular fibrillation (VF) and six cases of sudden understanding of the mechanism of ibrutinib-induced
cardiac deaths [55]. The median age of these patients cardiac toxicity is warranted.
6 C. P. S. TANG ET AL.

Management of patients with BTK inhibitor major hemorrhage in patients using warfarin [57].
induced AF The United States and European regulatory agencies
provide contradictory recommendations regarding
Balancing thrombosis versus bleeding risk
the concomitant use of ibrutinib and warfarin.
In the general population, the risk of cardioembolic Concomitant warfarin use is contraindicated by
stroke from AF may be estimated with the widely European Medicines Agency but is acceptable by FDA
used model CHA2DS2VASc. Contemporary guidelines [70,71]. Other forms of anticoagulation (most com-
recommend therapeutic anticoagulation for monly low-molecular-weight heparin) and antiplatelet
CHA2DS2VASc of 2 or greater [63]. However, as dis- agents have been used in trials with acceptable
cussed below, patients receiving ibrutinib are at a bleeding risk [67].
heightened bleeding risk due to the combination of Direct oral anticoagulants (DOAC) may be the pre-
antiplatelet effect of ibrutinib, the disease (CLL) itself, ferred therapy of choice due to the ease of administra-
baseline thrombocytopenia and concomitant anti- tion and reduced major and fatal bleeding compared
coagulant use. to warfarin. In a meta-analysis and systematic review
Platelets express both BTK and TEC which help to of clinical trials performed in noncancer settings,
Downloaded by [Imperial College London Library] at 04:49 14 September 2017

activate platelet aggregation by regulating phospho- DOAC significantly reduced the rate of major bleeding
lipase C!2 downstream of the collagen receptor glyco- (relative risk 0.72) and fatal bleeding (relative risk 0.53).
protein VI (GPVI) [64]. Ibrutinib possesses potent In the absence of similar trials performed in cancer set-
antiplatelet effects due to inhibition of collagen and tings, it may not be unreasonable to extrapolate this
von Willebrand factor-dependent platelet functions data to our patient population [72]. Caution must be
downstream of GPVI [65]. The degree of reduction of exercised though due to potential clinically significant
in vitro collagen-mediated platelet aggregation in drug–drug interactions. Ibrutinib is predominantly
blood from ibrutinib-treated patients was correlated metabolized by hepatic cytochrome P450 3A4
with the frequency of bleeding, which was reversible (CYP3A4) and to a lesser extent CYP2D6.
following addition of untreated platelets and drug ces- Coadministration of CYP3A perpetrators (i.e. CYP3A
sation [65,66]. inducers or inhibitors) can cause significant drug–drug
In clinical trials, ibrutinib was associated with interactions (DDI), which may alter the toxicity and
approximately 50% risk of bleeding, albeit the majority efficacy of the medications [73,74]. Even though both
being grade 1–2 bleeds consisting of petechiae and apixiban and rivaroxaban are substrates of CYP3A4,
contusion [4,6,26,28]. Major bleeding (grade 3) the clinical relevance of coadministration of both sub-
ranged from 1–10% in major trials (Table 4). Notably, strates is not established, unless there is possibility of
secondary analysis from two studies of single-agent enzyme saturation (which is uncommon for this group
ibrutinib trials (PCYC-1102 and RESONATE) showed of drugs). A personalized, risk-adapted approach
that anticoagulant (11%) and antiplatelet agents (34%) should be taken when selecting an anticoagulant, sup-
use were frequent amongst ibrutinib patients and that plemented by tools such as HAS-BLED (hypertension,
3% of this population had major bleeding events abnormal renal/liver function, stroke, bleeding history
[22,27,67]. Most of these events tended to occur early or predisposition, labile international normalized ratio
during treatment (within 3–6 months of therapy) and (INR)) for patients who were on warfarin or HAS-BED
led to drug discontinuation in 1% of patients [67]. for treatment naïve patients (hypertension, abnormal
However, Thompson et al. reported the risk of grade renal/liver function, stroke, bleeding history or predis-
3–4 bleeding in patients who are anticoagulated for position, elderly, drugs/alcohol) [75–77]. Factor Xa
ibrutinib-induced AF was much higher (14%). inhibitors may be the preferred choice of anticoagu-
Furthermore, CLL itself induces platelet dysfunction by lant in the elderly patient cohort with multiple comor-
impairing both adenosine diphosphate (ADP) and col- bidities. Conversely, it is feasible to continue warfarin
lagen-mediated platelet dysfunction [68]. for patients who were taking warfarin with stable INR
prior to commencement of ibrutinib [78]. Although
firm evidence is lacking, it may be reasonable to lower
Choice of anticoagulant
the dose of ibrutinib in those patients who develop
There is a paucity of safety data regarding the opti- bleeding manifestations while on concurrent anticoa-
mal choice of anticoagulation in ibrutinib patients gulation. Improved awareness of medications that may
who develop AF with elevated embolic stroke risk impact the CYP3A4, especially when anticoagulation is
[69]. Most of the trials involving ibrutinib prohibited taken in combination is needed. Alarmingly, a recent
concurrent use of warfarin following observations of report showed that a significant proportion of patients
CARDIOTOXICITY OF BTK 7

(64%) on ibrutinib were on concurrent medications Future directions


that may alter its metabolism [79]. Concurrent adminis-
The potency of ibrutinib in treating B-cell malignancies
tration of CYP3A4 inhibitors in this instance may lead
is unquestionable. However, the risk of AF is not negli-
to clinically significant bleeding.
gible and complicated by management issues such as
For patients with intermediate stroke risk
balancing bleeding versus clotting risk and pharmaco-
(CHA2DS2VASc of 1), the addition of antiplatelet agent
logical drug interactions. Notwithstanding the net
for younger patients with few comorbidites may not
benefit of therapy, the critical issues of AF need to be
be unreasonable as even though ibrutinib has antipla-
addressed ideally with general consensus guidelines.
telet properties, it is unclear if it confers protective
Closer collaboration between hematologists and cardi-
role against cardioembolic stroke. Further studies are
required to enable physicians to make informed deci- ologists is needed in both clinical trial designs and
sions regarding anticoagulation in patients who are on adjudicating cardiovascular end points. A risk-adapted
ibrutinib. approach needs to be employed in therapy selection,
especially in the front-line setting taking into consider-
ation patient demographics, cardiovascular risk factors
Downloaded by [Imperial College London Library] at 04:49 14 September 2017

Management of arrhythmia and heart failure and nature of disease. Several second-generation BTK
The complexity of management of ibrutinib-induced inhibitors are also on the horizon with the potential
AF and heart failure is further compounded by the promise of improved efficacy and safety profile.
lack of evidence-based guidelines regarding the (1)
utility and method of surveillance, (2) choice between Acalabrutinib (ACP-196)
pharmacological treatment or interventional therapy
Similar to ibrutinib, acalabrutinib is an irreversible BTK
such as direct current cardioversion and (3) safety and
inhibitor that binds covalently to the cysteine-481 resi-
efficacy regarding ibrutinib cessation or dose reduc-
due. Contrary to ibrutinib, it has enhanced selectivity
tion. Thompson et al. retrospective study found that in
to BTK with potential improvement of potency and
the majority of cases, AF persisted despite stopping or
less off-target activity on EFGR, TEC and ITK, which
reducing the dose of ibrutinib [52]. Historically, both
may result in less untoward side effects such as bleed-
rate control and rhythm control confer similar efficacy
ing, severe diarrhea and rash, and possibly AF [83,84].
in the reduction of mortality, heart failure and rate of
In a phase 1–2 trial involving 61 heavily relapsed or
hospitalizations from AF in the noncancer patient
refractory patients (median of 3 previous therapies)
population [80,81]. We usually select rate control with
and majority of high risk patients (31% had TP53 dys-
beta blockers for patients with ibrutinib-induced AF, as
regulation and 75% had unmutated IGHV), the overall
the majority of antiarrhythmic agents used in pharma-
response rate was 95% after a median follow-up of
cological cardioversion are pro-arrhythmic in nature.
14.3 months. Encouragingly, the ORR for the subset of
Secondly, pertinent drug interactions occur between
patients with chromosome 17p13.1 deletion was
ibrutinib and several antiarrhythmic agents (amiodar- 100%. There were no reports of dose-limiting toxicities,
aone, verapamil and diltiazem) via CYP3A4 hepatic major hemorrhage and AF. Severe bleeding occurred
metabolism. Pharmacovigilance among physicians in less than 2% of patients. Only 19% of patients
again is mandated. Electrical cardioversion may be experienced grade 3–4 toxicities (most commonly
prudent for symptomatic persistent or permanent AF hypertension). The most common side effects
failing rate control with beta blockers. Inevitably, there were headache, diarrhea and weight gain [85]. No
are patients in whom we need to cease or interrupt cases of AF have been publically reported for acalabru-
their ibrutinib treatment when they develop grade tinib to date.
3 AF which fails to respond to pharmacological or
interventional therapy. Barr et al showed that sus-
tained treatment adherence was associated with better BGB-3111
PFS, higher ORR and a trend toward improved ORR, BGB-3111 is a second-generation BTKi which is equipo-
irrespective of del 17p or TP53 mutation. Treatment tent against BTK compared to ibrutinib with less
interruption for just 1 week was associated with inhibitory effect on EGFR, TEC, ITK, HER 2 and JAK 3. In
increased PFS events [82]. This growing group of a phase-1 study involving 45 patients with R/R CLL, of
patients is particularly difficult to manage due to which 15% had del 17p, only 1 case of AF (grade 2)
decreased ibrutinib efficacy and a relative paucity of and major bleeding (grade 3 purpura) was reported
alternate treatment options [48,51]. out of the 29 patients who were available for analysis.
8 C. P. S. TANG ET AL.

The most common side effects were mild (grade 1/2) high response with less toxicity in longer-term follow-
and comprised of bruising/bleeding, diarrhea and up reports.
upper respiratory infections. After a median follow-up
of 7.5 months, the ORR was 90% and there was no
Conclusions
reported disease progression or Richter’s transform-
ation [86]. This drug has also demonstrated high clin- Ibrutinib is undoubtedly an effective therapy for B-cell
ical activity in WM. In a recent phase 1–2 study malignancies but results in an increased rate of AF
involving 24 patients who were either treatment naïve with consequences of potentially excessive bleeding
or R/R, the ORR was 92%, with 33% achieving very risk and discontinuation of treatment. The manage-
good partial response (33%) after a median follow-up ment of this cohort of patients is truly an area of
of 7.6 months. There were two cases of AF (grade 1–2) unmet needs, which mandates urgent exploration in
and no reports of grade 3 bleeding [87]. future prospective studies involving both hematolo-
gists and cardiologists especially since it will be used
increasingly in the upfront setting. Vigilance is needed
ONO/GS 4059 among treating physicians with regards to the nuances
Downloaded by [Imperial College London Library] at 04:49 14 September 2017

ONO/GS 4059 is another second-generation BTK inhibi- of the treatment for these patients.
tor that forms an irreversible covalent bond to a cyst-
eine residue at the BTK active site. It has high oral Potential conflict of interest: Disclosure forms pro-
bioavailability with high selectivity for BTK compared vided by the authors are available with the full text of
to ibrutinib but has less inhibitory effect on EGFR and this article online at https://doi.org/ 10.1080/10428194.
ITK [88]. Walter et al. performed a phase 1–2 single- 2017.1375110.
arm study with ONO/GS 4059 on 28 R/R high genetic
risk (del 17p 36%; TP53 mutation 52%; unmutated References
IGHV 84%) CLL patients. Anticoagulation was allowed
[1] Herman SEM, Gordon AL, Hertlein E, et al. Bruton
for this study; 6/28 patients were on anticoagulant tyrosine kinase represents a promising therapeutic tar-
therapy. At a median follow-up of 32.5 months, 39.3% get for treatment of chronic lymphocytic leukemia
(11) of patients dropped out of the study (3 due to and is effectively targeted by PCI-32765. Blood. 2011;
disease progression; 3 due to death and 4 due to tox- 117:6287–6296.
icities). The estimated median progression-free survival [2] Honigberg LA, Smith AM, Sirisawad M, et al. The
Bruton tyrosine kinase inhibitor PCI-32765 blocks B-
was 38.5 months and median overall survival was 44.9 cell activation and is efficacious in models of auto-
months. Overall response rate was 96%. There were no immune disease and B-cell malignancy. Proc Natl
new treatment emergent side effects on the updated Acad Sci USA. 2010;107:13075–13080.
data. The use of anticoagulants in this cohort was not [3] Cheng S, Ma J, Guo A, et al. BTK inhibition targets in
associated with increased risk of bleeding. There was 1 vivo CLL proliferation through its effects on B-cell
receptor signaling activity. Leukaemia. 2014;28:649–657.
case of grade-3 bleeding (spontaneous psoas hema- [4] Byrd JC, Furman RR, Coutre SE, et al. Three-year fol-
toma), but this patient was not on anticoagulation. low-up of treatment-naive and previously treated
One case of AF was reported, but it was provoked by patients with CLL and SLL receiving single-agent ibru-
pneumonia. Overall, the treatment was well tolerated tinib. Blood. 2015;125:2497–2506.
with the most common side effects being bruising [5] Burger JA, Tedeschi A, Barr PM, et al. Ibrutinib as ini-
tial therapy for patients with chronic lymphocytic leu-
(35.7% all grades), neutropenia (35.7% all grades) and kemia. N Engl J Med. 2015;373:2425–2437.
anemia (32.1%) [89]. [6] Wang ML, Blum KA, Martin P, et al. Long-term follow-
Clearly, it is still premature to determine if these up of MCL patients treated with single-agent ibruti-
newer BTK inhibitors are comparable if not more nib: updated safety and efficacy results. Blood. 2015;
superior to ibrutinib with better safety profiles. It is 126:739–745.
[7] Treon SP, Tripsas CK, Meid K, et al. Ibrutinib in
hoped that the ongoing ELEVATE trial, which is a previously treated Waldenstro €m’s macroglobulinemia.
phase-3 noninferiority trial comparing ibrutinib and N Engl J Med. 2015;372:1430–1440.
acalabrutinib in treating high-risk, relapsed/refractory [8] Gertz MA. Waldenstro €m macroglobulinemia: 2017
CLL patients will shed more light in this area in the update on diagnosis, risk stratification, and manage-
very near future [90]. Currently, in the absence of ment. Am J Hematol. 2017;92:209–217.
[9] Ponader S, Chen SS, Buggy JJ, et al. The Bruton tyro-
head-to-head trial outcomes, it may be difficult to sine kinase inhibitor PCI-32765 thwarts chronic
draw precise conclusions, but it will be tantalizing to lymphocytic leukemia cell survival and tissue homing
see if these newer agents deliver the same durable in vitro and in vivo. Blood. 2012;119:1182–1189.
CARDIOTOXICITY OF BTK 9

[10] de Rooij MFM, Kuil A, Geest CR, et al. The clinically lymphocytic leukemia/small lymphocytic leukemia.
active BTK inhibitor PCI-32765 targets B-cell receptor- Blood. 2016;128:234.
and chemokine-controlled adhesion and migration in [26] Farooqui MZH, Valdez J, Martyr S, et al. Ibrutinib for
chronic lymphocytic leukemia. Blood. 2012;119: previously untreated and relapsed or refractory
2590–2594. chronic lymphocytic leukaemia with TP53 aberrations:
[11] Herman SEM, Mustafa RZ, Gyamfi JA, et al. Ibrutinib a phase 2, single-arm trial. Lancet Oncol. 2015;16:
inhibits BCR and NF- B signaling and reduces tumor 169–176.
proliferation in tissue-resident cells of patients with [27] Byrd JC, Brown JR, O'Brien S, et al. Ibrutinib versus
CLL. Blood. 2014;123:3286–3295. ofatumumab in previously treated chronic lymphoid
[12] Jemal A, Siegel R, Ward E, et al. Cancer statistics, leukemia. N Engl J Med. 2014;371:213–223.
2007. CA Cancer J Clin. 2007;57:43–66. [28] Jaglowski SM, Jones JA, Nagar V, et al. Safety and
[13] Rozman C, Montserrat E. Chronic lymphocytic leukae- activity of BTK inhibitor ibrutinib combined with ofa-
mia. N Engl J Med. 1995;333:1515. tumumab in chronic lymphocytic leukemia: a phase
[14] Woyach JA, Johnson AJ, Byrd JC. The B-cell receptor 1b/2 study. Blood. 2015;126:842–850.
signaling pathway as a therapeutic target in CLL. [29] Burger JA, Keating MJ, Wierda WG, et al. Safety and
Blood. 2012;120:1175–1184. activity of ibrutinib plus rituximab for patients with
[15] €
Minden MD-V, Ubelhart R, Schneider D, et al. Chronic high-risk chronic lymphocytic leukaemia: a single-arm,
Downloaded by [Imperial College London Library] at 04:49 14 September 2017

lymphocytic leukaemia is driven by antigen-independ- phase 2 study. Lancet Oncol. 2014;15:1090–1099.


ent cell-autonomous signalling. Nature. 2012;489: [30] Rossi D, Terzi-di-Bergamo L, De Paoli L, et al.
309–312. Molecular prediction of durable remission after first-
[16] Mohamed AJ, Yu L, B€ackesjo € C-M, et al. Bruton’s tyro- line fludarabine-cyclophosphamide-rituximab in
sine kinase (Btk): function, regulation, and transform- chronic lymphocytic leukemia. Blood. 2015;126:
ation with special emphasis on the PH domain. 1921–1924.
Immunol Rev. 2009;228:58–73. [31] Zenz T, Eichhorst B, Busch R, et al. TP53 mutation and
[17] Takata M, Kurosaki T. A role for Bruton's tyrosine kin-
survival in chronic lymphocytic leukemia. J Clin Oncol.
ase in B cell antigen receptor-mediated activation of
2010;28:4473–4479.
phospholipase C-gamma 2. J Exp Med. 1996;184:
[32] Fischer K, Bahlo J, Fink AM, et al. Long-term remis-
31–40.
sions after FCR chemoimmunotherapy in previously
[18] Burger JA, Chiorazzi N. B cell receptor signaling in
untreated patients with CLL: updated results of the
chronic lymphocytic leukemia. Trends Immunol. 2013;
CLL8 trial. Blood. 2016;127:208–215.
34:592–601.
[33] Thompson PA, Wierda WG. Eliminating minimal
[19] Okkenhaug K, Vanhaesebroeck B. PI3K in lymphocyte
residual disease as a therapeutic end point: working
development, differentiation and activation. Nat Rev
toward cure for patients with CLL. Blood. 2016;127:
Immunol. 2003;3:317–330.
279–286.
[20] Hoellenriegel J, Meadows SA, Sivina M, et al. The
[34] Imbruvica prescribing information. 2017. https://www.
phosphoinositide 30 -kinase delta inhibitor, CAL-101,
imbruvica.comdocslibrariesproviderdefault-document-
inhibits B-cell receptor signaling and chemokine net-
works in chronic lymphocytic leukemia. Blood. libraryprescribinginformation.pdf.
2011;118:3603–3612. [35] Wang ML, Rule S, Martin P, et al. Targeting BTK with
[21] Hacken ten E, Burger JA. Microenvironment interac- ibrutinib in relapsed or refractory mantle-cell lymph-
tions and B-cell receptor signaling in chronic lympho- oma. N Engl J Med. 2013;369:507–516.
cytic leukemia: implications for disease pathogenesis [36] Dreyling M, Jurczak W, Jerkeman M, et al. Ibrutinib
and treatment. BBA - Mol Cell Res. 2016;1863: versus temsirolimus in patients with relapsed or
401–413. refractory mantle-cell lymphoma: an international,
[22] Byrd JC, Furman RR, Coutre SE, et al. Targeting BTK randomised, open-label, phase 3 study. Lancet. 2016;
with ibrutinib in relapsed chronic lymphocytic leuke- 387:770–778.
mia. N Engl J Med. 2013;369:32–42. [37] Owen RG, Kyle RA, Stone MJ, et al. Response assess-
[23] Chanan-Khan A, Cramer P, Demirkan F, et al. Ibrutinib ment in Waldenstro €m macroglobulinaemia: update
combined with bendamustine and rituximab com- from the VIth International Workshop. Br J Haematol.
pared with placebo, bendamustine, and rituximab for 2012;160:171–176.
previously treated chronic lymphocytic leukaemia or [38] Treon SP, Xu L, Yang G, et al. MYD88 L265P somatic
small lymphocytic lymphoma (HELIOS): a randomised, mutation in Waldenstro €m's macroglobulinemia. N
double-blind, phase 3 study. Lancet Oncol. 2016;17: Engl J Med. 2012;367:826–833.
200–211. [39] Xu L, Hunter ZR, Yang G, et al. MYD88 L265P in
[24] O'Brien S, Jones JA, Coutre SE, et al. Ibrutinib for Waldenstro €m macroglobulinemia, immunoglobulin M
patients with relapsed or refractory chronic lympho- monoclonal gammopathy, and other B-cell lympho-
cytic leukaemia with 17p deletion (RESONATE-17): a proliferative disorders using conventional and quanti-
phase 2, open-label, multicentre study. Lancet Oncol. tative allele-specific polymerase chain reaction. Blood.
2016;17(10):1409–1418. 2013;121:2051–2058.
[25] Barr P. Updated efficacy and safety from the phase 3 [40] Dimopoulos MA, Trotman J, Tedeschi A, et al.
resonate-2 study: ibrutinib as first-line treatment Ibrutinib for patients with rituximab-refractory
option in patients 65 years and older with chronic Waldenstro €m's macroglobulinaemia (iNNOVATE): an
10 C. P. S. TANG ET AL.

open-label substudy of an international, multicentre, through inhibition of cardiac PI3K-Akt signaling.


phase 3 trial. Lancet Oncol. 2017;18:241–250. Blood. 2014;124:3829–3830.
[41] Stewart DM, Tian L, Nelson DL. The clinical spectrum [58] Qiu Y, Kung HJ. Signaling network of the Btk family
of bruton’s agammaglobulinemia. Curr Allergy Asthma kinases. Oncogene. 2000;19:5651–5661.
Rep. 2001;1:558–565. [59] Pretorius L, Du XJ, Woodcock EA, et al. Reduced phos-
[42] Leong DP, Caron F, Hillis C, et al. The risk of atrial fib- phoinositide 3-kinase (p110a) activation increases the
rillation with ibrutinib use: a systematic review and susceptibility to atrial fibrillation. Am J Pathol. 2010;
meta-analysis. Blood. 2016;128:138–140. 175:998–1009.
[43] Shanafelt TD, Parikh SA, Noseworthy PA, et al. Atrial [60] Yang T, Moleslehi J, Roden DM. Proarrhythmic effects
fibrillation in patients with chronic lymphocytic leuke- of ibrutinib, a clinically approved inhibitor of Bruton’S
mia (CLL). Leuk Lymphoma. 2017;58:1630–1639. tyrosine kinase (BTK) used in cancer therapy.
[44] Farooqui M, Wiestner A, Valdez J. Atrial Fibrillation in Circulation. 2015;132(Supp 3):A14587.
CLL/SLL Patients on Ibrutinib. 2015. [61] Nair KS, Cheson B. The role of idelalisib in the treat-
[45] Coutre SE, Furman RR, Flinn IW, et al. Extended treat- ment of relapsed and refractory chronic lymphocytic
ment with single-agent ibrutinib at the 420 mg dose leukemia. Ther Adv Hematol. 2016;7:69–84.
leads to durable responses in chronic lymphocytic [62] Chung C, Lee R. Ibrutinib, obinutuzumab, idelalisib,
leukemia/small lymphocytic lymphoma. Clin Cancer and beyond: review of novel and evolving therapies
Downloaded by [Imperial College London Library] at 04:49 14 September 2017

Res. 2017;23:1149–1155. for chronic lymphocytic leukemia. Pharmacotherapy.


[46] Jain P, Keating MJ, Wierda WG, et al. Long-term fol- 2014;34:1298–1316.
low-up of treatment with ibrutinib and rituximab in [63] January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/
patients with high-risk chronic lymphocytic leukemia. HRS guideline for the management of patients with
Clin Cancer Res. 2016;23:2154–2158. atrial fibrillation: a report of the American College of
[47] Mato AR, Nabhan C, Barr PM, et al. Outcomes of CLL Cardiology/American Heart Association Task Force on
patients treated with sequential kinase inhibitor ther- Practice Guidelines and the Heart Rhythm Society.
apy: a real world experience. Blood. 2016;128: J Am Coll Cardiol. 2014;64(21):e1–76.
2199–2205. [64] Atkinson BT. Tec regulates platelet activation by GPVI
[48] Jain P, Thompson PA, Keating M, et al. Long-term out- in the absence of Btk. Blood. 2003;102:3592–3599.
comes for patients with chronic lymphocytic leukemia [65] Levade M, David E, Garcia C, et al. Ibrutinib treatment
who discontinue ibrutinib. Cancer. affects collagen and von Willebrand factor-dependent
2017;123:2268–2273. platelet functions. Blood. 2014;124:3991–3995.
[49] Maddocks K, Jones JA. Bruton tyrosine kinase inhib- [66] Kamel S, Horton L, Ysebaert L, et al. Ibrutinib inhibits
ition in chronic lymphocytic leukemia. Semin Oncol. collagen-mediated but not ADP-mediated platelet
2016;43:251–259. aggregation. Leukaemia. 2015;29:783–787.
[50] Jain P, Keating M, Wierda W, et al. Outcomes of [67] Jones JA, Hillmen P, Coutre S, et al. Use of anticoagu-
patients with chronic lymphocytic leukemia after dis- lants and antiplatelet in patients with chronic lympho-
continuing ibrutinib. Blood. 2015;125:2062–2067. cytic leukaemia treated with single-agent ibrutinib. Br
[51] Thompson PA, Cymbalista F, Keating M, Tam CS. The J Haematol. 2017;102:3516–3592.
impact of atrial fibrillation on subsequent survival of [68] Lipsky AH, Farooqui MZH, Tian X, et al. Incidence and
patients receiving ibrutinib as treatment of chronic risk factors of bleeding-related adverse events in
lymphocytic leukemia (CLL): An International Study. patients with chronic lymphocytic leukemia treated
2016. with ibrutinib. Haematologica. 2015;100:1571–1578.
[52] Thompson PA, Levy V, Tam CS, et al. Atrial fibrillation [69] Shatzel JJ, Olson SR, Tao DL, et al. Ibrutinib-associated
in CLL patients treated with ibrutinib. An international bleeding: pathogenesis, management and risk reduc-
retrospective study. Br J Haematol. 2016;175:462–466. tion strategies. J Thromb Haemost. 2017;32(Suppl. 1):
[53] Fauchier L, Villejoubert O, Clementy N, et al. Causes 1830.
of death and influencing factors in patients with atrial [70] FDA. 2015. [Internet]. http://www.accessdata.fda.gov-
fibrillation. Am J Med. 2016;129:1278–1287. drugsatfdadocslabel-slbl.pdf. 2015;1–26.
[54] Mato A, Carver J. Left Atrial Abnormality as a [71] European Medicines Agency. http://www.ema.europa.
Predictor of ibrutinib-associated atrial fibrillation in eudocsenGBdocumentlibraryEPAR-
patients with chronic lymphocytic leukemia. XVII ProductInformationhumanWC.pdf. 2017;1–37.
International Workshop on Chronic Lymphocytic [72] Chai-Adisaksopha C, Crowther M, Isayama T, et al. The
Leukaemia. 2017. impact of bleeding complications in patients receiving
[55] Lampson BL, Yu L, Glynn RJ, et al. Ventricular arrhyth- target-specific oral anticoagulants: a systematic review
mias and sudden death in patients taking ibrutinib. and meta-analysis. Blood. 2014;124:2450–2458.
Blood. 2017;129:2581–2584. [73] Scheers E, Leclercq L, de Jong J, et al. Absorption,
[56] Tomcsanyi J, Nenyei Z, Matrai Z, et al. Ibrutinib, an metabolism, and excretion of oral 14C radiolabeled
approved tyrosine kinase inhibitor as a potential ibrutinib: an open-label, phase I, single-dose study in
cause of recurrent polymorphic ventricular tachycar- healthy men. Drug Metab. Dispos. 2015;43:289–297.
dia. JACC: Clin Electrophysiol. 2016;2:847–849. [74] de Jong J, Skee D, Murphy J, et al. Effect of CYP3A
[57] McMullen JR, Boey EJH, Ooi JYY, et al. Ibrutinib perpetrators on ibrutinib exposure in healthy partici-
increases the risk of atrial fibrillation, potentially pants. Pharmacol Res Perspect. 2015;3:e00156.
CARDIOTOXICITY OF BTK 11

[75] Poli D, Antonucci E, Pengo V, et al. Comparison of [83] Harrington B, Johnson AJ, Gulrajani M, et al. ACP-196
HAS-BLED and HAS-BED versus CHADS2 and is a second generation inhibitor of bruton tyrosine
CHA2DS2VASC stroke and bleeding scores in patients kinase (BTK) with enhanced target specificity. Blood.
with atrial fibrillation. Am J Cardiol. 2017;119: 2015;126:2908.
1012–1016. [84] Wu J, Zhang M, Liu D. Acalabrutinib (ACP-196): a
[76] Lip GYH, Lane DA. Bleeding risk assessment in atrial selective second-generation BTK inhibitor. J Hematol
fibrillation: observations on the use and misuse of Oncol. 2016;9:21.
bleeding risk scores. J Thromb Haemost. 2016;14: [85] Byrd JC, Harrington B, O'Brien S, et al. Acalabrutinib
1711–1714. (ACP-196) in relapsed chronic lymphocytic leukemia.
[77] Lip GYH. Recommendations for thromboprophylaxis N Engl J Med. 2016;374:323–332.
in the 2012 focused update of the ESC guidelines on [86] Tam CS, Grigg AP, Opat S. The BTK inhibitor, Bgb-
atrial fibrillation: a commentary. J Thromb Haemost. 3111, is safe, tolerable, and highly active in patients
with relapsed/refractory B-cell malignancies: initial
2013;11:615–626.
report of a phase 1 first-in-human trial. Blood. 2015;
[78] Chai KL, Rowan G, Seymour JF, et al. Practical recom-
126:832.
mendations for the choice of anticoagulants in the
[87] Tam CS, Seymour J, Grigg AP. High major response
management of patients with atrial fibrillation on
rate, including very good partial responses (VGPR), in
ibrutinib. Leuk. Lymphoma. 2017;128:1–4.
Downloaded by [Imperial College London Library] at 04:49 14 September 2017

patients (pts) with Waldenstrom macroglobulinemia


[79] Finnes HD, Chaffee KG, Call TG, et al.
(WM) treated with the highly specific BTK inhibitor
Pharmacovigilance during ibrutinib therapy for
Bgb-3111: expansion phase results from an ongoing
chronic lymphocytic leukemia (CLL)/small lymphocytic phase I study. Blood. 2016;128:1216.
lymphoma (SLL) in routine clinical practice. Leuk. [88] Walter HS, Rule SA, Dyer MJS, et al. A phase 1 clinical
Lymphoma. 2017;58:1376–1383. trial of the selective BTK inhibitor ONO/GS-4059 in
[80] Roy D, Talajic M, Nattel S, et al. Rhythm control versus relapsed and refractory mature B-cell malignancies.
rate control for atrial fibrillation and heart failure. N Blood. 2016;127:411–419.
Engl J Med. 2008;358:2667–2677. [89] Walter HS, Jayne S, Rule SA, et al. Long-term follow-
[81] Chinitz JS, Halperin JL, Reddy VY, et al. Rate or rhythm up of patients with CLL treated with the selective
control for atrial fibrillation: update and controversies. Bruton's tyrosine kinase inhibitor ONO/GS-4059.
Am J Med. 2012;125:1049–1056. Blood. 2017;129:2808–2810.
[82] Barr PM, Brown JR, Hillmen P, et al. Impact of ibruti- [90] Elevate CLL R/R: Study of Acalabrutinib (ACP-196)
nib dose adherence on therapeutic efficacy in patients Versus Ibrutinib in Previously Treated Subjects With
with previously treated CLL/SLL. Blood. 2017;129: High Risk Chronic Lymphocytic Leukemia. http:sclini-
2612–2615. caltrials.govctshowNCT. 2017.

You might also like