Sunteți pe pagina 1din 6

Thrombosis Research 133 (2014) 10231028

Contents lists available at ScienceDirect

Thrombosis Research
journal homepage: www.elsevier.com/locate/thromres

Regular Article

Subcutaneous Enoxaparin for Therapeutic Anticoagulation in


Hemodialysis Patients
Tiffany K. Pon a,, William E. Dager b,c,1, A. Joshua Roberts d,e,2, Richard H. White f,3
a

Clinical Pharmacy, University of California, San Francisco, CA, 2315 Stockton Blvd, Sacramento, CA 95815, USA
University of California, Davis Medical Center, University of California, San Francisco, CA, USA
University of California, Davis School of Medicine, Touro Vallejo School of Pharmacy, 2315 Stockton Blvd, Sacramento, CA 95815, USA
d
University of California, Davis Medical Center, Clinical Pharmacy, University of California, San Francisco, CA, USA
e
University of California, Davis School of Medicine, 2315 Stockton Blvd, Sacramento, CA 95815, USA
f
Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA, USA
b
c

a r t i c l e

i n f o

Article history:
Received 6 January 2014
Received in revised form 5 March 2014
Accepted 18 March 2014
Available online 24 March 2014
Keywords:
Heparin
Low-Molecular-Weight Heparin
Enoxaparin
Renal Dialysis
Thromboembolism
Hemorrhage

a b s t r a c t
Background: Information regarding dosing of low-molecular-weight heparins (LMWH) for therapeutic anticoagulation in hemodialysis (HD) patients is limited. The aim of this study was to retrospectively compare the safety
and efcacy of enoxaparin versus unfractionated heparin (UFH) for therapeutic anticoagulation in HD patients.
Materials and Methods: This retrospective chart review evaluated HD patients treated with subcutaneous enoxaparin
that were matched based on the indication for anticoagulation with patients treated with intravenous UFH
to achieve therapeutic anticoagulation. Primary outcome measures included 30-day incidence of thromboembolic
events and major bleeding. Secondary outcomes included rehospitalization within 30 days, length of stay, and
mortality.
Results: One hundred sixty-four patients were evaluated, 82 in each group. The average daily dose of enoxaparin
used to target therapeutic levels was 0.7 0.2 mg/kg/day (range = 0.4-1). Comparing enoxaparin to UFH,
there was no signicant difference in major bleeding (6.1% vs 11%, p = 0.4) or thromboembolism (0% vs 2.4%,
p = 0.5). Hospital length of stay was shorter in the enoxaparin group (20 53.8 vs 28.9 44.5 days, p = 0.02);
there was no signicant difference between groups in mortality or readmission. Adjusting for risk factors for bleeding there was a slight but statistically non-signicant difference between enoxaparin versus UFH (OR = 0.77, 95%CI:
0.2-3.5, p = 0.73).
Conclusions: These ndings suggest that therapeutic dosing of enoxaparin, in doses that ranged from 0.41 mg/kg/day, was as safe as intravenous UFH in providing therapeutic anticoagulation in stable patients requiring
chronic hemodialysis.
2014 Elsevier Ltd. All rights reserved.

Introduction

Abbreviations: IV, intravenous; UFH, unfractionated heparin; LMWH, low-molecularweight heparins; VTE, venous thromboembolism; ACS, acute coronary syndrome; CrCl,
creatinine clearance; UCDMC, University of California, Davis Medical Center; ISTH,
International Society on Thrombosis and Haemostasis; SAS, Statistical Analysis
Software; CAD, coronary artery disease; CVA, cerebrovascular accident; IHD, intermittent
hemodialysis; CRRT, continuous renal replacement; SLEDD, slow-extended daily dialysis;
PD, peritoneal dialysis.
Research Location: University of California, Davis Medical Center, 2315 Stockton Blvd,
Sacramento, CA 95815.
Tel.: +1 916 734 7726; fax: +1 916 703 4008.
E-mail addresses: tiffany.pon@ucsf.edu (T.K. Pon), william.dager@ucdmc.ucdavis.edu
(W.E. Dager), aaron.roberts@ucdmc.ucdavis.edu (A.J. Roberts), rhwhite@ucdavis.edu
(R.H. White).
1
Tel.: +1 916 703 4025; fax: +1 916 703 5618.
2
Tel.: +1 916 703 4024; fax: +1 916 703 5618.
3
Te.: +1 916 734 7005; fax: +1 916 734 2732.

http://dx.doi.org/10.1016/j.thromres.2014.03.036
0049-3848/ 2014 Elsevier Ltd. All rights reserved.

Options for full-dose therapeutic anticoagulation in patients with


chronic renal failure who require hemodialysis (HD) are limited, with
few alternatives to the gold-standards of intravenous (IV) unfractionated
heparin (UFH) and oral vitamin K antagonists (e.g. warfarin) [1,2].
Nevertheless there are clinical situations when subcutaneously injected
low-molecular-weight heparins (LMWH) might be potentially advantageous. These include bridging patients onto or off of warfarin (including
perioperative bridging), treating patients with absent or poor venous
access, and treating cancer patients with acute venous thromboembolism (VTE) [3]. Although these scenarios are not common, they are not
rare. A comparison of HD patients treated with UFH versus LMWH
would be useful to better dene the incidence of bleeding and thrombotic
outcomes in patients managed with these drugs.
Because LMWH have reduced renal clearance, large volumes of
distribution, and longer half-lives than UFH, the assumed risk of using

1024

T.K. Pon et al. / Thrombosis Research 133 (2014) 10231028

these drugs in hemodialysis patients is accumulation, excessive


anticoagulation and subsequent bleeding [48]. Current practice guidelines also state LMWH should be avoided in HD patients; however,
there have been no prospective studies that have directly assessed
clinical outcomes of LMWH treatment in this population. Enoxaparin
(Lovenox, Bridgewater, New Jersey, United States of America) has
been shown to accumulate in patients treated with therapeutic doses
(1.0-1.25 mg/kg) given subcutaneously every 12 hours for non-STsegment elevation acute coronary syndrome (ACS), with a strong linear
relationship between the creatinine clearance (CrCl) and drug clearance
[5]. In this study by Becker et al., only 11 of 445 (2.5%) patients in their cohort had impaired renal function, dened as CrCl less than 40 mL/min.
Major hemorrhage, a secondary outcome of the study, occurred in 27 of
445 (6.1%) patients, but it is unclear how many of these patients were
in the impaired renal function group. Importantly, the enoxaparin dose
was not reduced or adjusted for decreased renal function.
A recent study evaluated the use of standard therapeutic doses of
dalteparin and tinzaparin for perioperative bridging in HD patients
and measured trough anti-Xa levels 2024 hours after dose administration [8]. Patients in this study had chronic renal failure and were receiving intermittent HD three times per week. This study documented
accumulation of tinzaparin and dalteparin at non-adjusted therapeutic
doses in these HD patients; however, the trial was neither designed
nor powered to make any inferences about the safety or efcacy of
treatment using these LMWH preparations, and the authors did not
report any clinical outcomes. Routine measurment of plasma anti-Xa
levels in HD patients or patients with severe renal insufciency treated
with a LMWH has not been validated as a useful or reliable parameter
for monitoring these patients [9].
Use of LMWH to prevent HD circuit thrombosis has been studied,
and these drugs are commonly used in HD centers [10]. Reports indicate
that use of enoxaparin is safe and effective when single lower doses of
0.4 to 0.7 mg/kg are delivered intravenously prior to the HD session
[7,1116]. In a study comparing enoxaparin to regular UFH, a modied
enoxaparin dose of 0.7 mg/kg IV prior to dialysis was shown to be
effective in maintaining circuit patency and was associated with a low
incidence of signicant bleeding; however, use of a LMWH for this indication remains an off-label practice [4,17].
Seeking alternative parenteral anticoagulation, physicians at the
University of California, Davis Medical Center (UCDMC) consulted
with the inpatient anticoagulation service and began treating select
HD patients with a modied, lower dose of subcutaneous enoxaparin
ranging 0.4-1 mg/kg daily. Actual dose selection was based principally
on assessment of the risk of thrombosis versus the risk of bleeding.
This lower adjusted treatment dose was used for several different
indications including acute VTE, bridging therapy, VTE prophylaxis following orthopedic surgery or multi-trauma, ACS, bridging for cardiac
valve replacement, stroke prevention for atrial brillation/utter,
hypercoagulable state, and cardioversion/ablation procedures.
Given the paucity of studies in medical literature that have rigorously
evaluated therapeutic dosing of enoxaparin (Lovenox) in HD patients,
the aim of this retrospective chart review was to compare the safety
and efcacy of therapeutic subcutaneous enoxaparin versus continuous
intravenous UFH in patients receiving various forms of HD (e.g. intermittent hemodialysis, continuous renal replacement therapy, slow extended
daily dialysis, and ultraltration). The primary efcacy endpoint was
30-day thromboembolic event and the primary safety endpoint was
the 30-day incidence of major bleeding.

HD patients. Patients were included in the study if they were age


18 years or older, required chronic or acute HD, and received at least
one dose of enoxaparin or were started on an UFH continuous infusion.
Consecutive patients receiving enoxaparin were matched 1:1 with randomly selected patients treated with IV UFH, based on the indication for
anticoagulation. Indications included the following: acute VTE, bridging
therapy, VTE prophylaxis following orthopedic surgery or multi-trauma,
ACS, bridging for cardiac valve replacement, stroke prevention for atrial
brillation/utter, hypercoagulable state, and cardioversion/ablation
procedures. Patients were excluded if they were treated with a LMWH
other than enoxaparin, received only prophylactic doses of anticoagulation (e.g. enoxaparin 30 mg daily), did not meet matching
criteria, or if they had incomplete medical records. The study was
approved by the UCDMC Institutional Review Board and requirement
for informed consent was waived.
Bleeding Risk Assessment and Monitoring
During chart review, patients were retrospectively assessed for
baseline bleeding risk based on the HAS-BLED scoring system [18].
Anti-factor Xa levels were not collected as they were not drawn during
hospitalization; patients receiving enoxaparin were monitored with
complete blood counts (CBC) and clinical signs and symptoms of bleeding. Patients receiving UFH were monitored using activated partial
thromboplastin time (APTT) in addition to CBC and clinical signs and
symptoms of bleeding. Baseline international normalized ratios (INR)
were also collected.
Outcome Measures
The primary efcacy endpoint was 30-day incidence of a symptomatic thromboembolic event. The primary safety endpoint was 30-day
incidence of International Society on Thrombosis and Haemostasis
(ISTH)-dened major bleeding, which is fatal bleeding and/or symptomatic bleeding in a critical organ or area (e.g. intracranial, intraspinal,
intraocular, retroperitoneal, intraarticular or pericardial, or intramuscular with compartment syndrome) and/or bleeding causing a fall in
hemoglobin level of 2 g/dL or more, or bleeding leading to transfusion
of two or more units of packed red blood cells [19]. Secondary outcomes
included the 30-day incidence of readmission for any reason, 30-day
all-cause mortality, and hospital length of stay. Readmission and
mortality as reported in the UCDMC electronic health record (EHR)
were the only late outcome measures; all other endpoints occurred
during hospitalization.
Statistical Analysis
A sample size calculation to ensure that a 15% difference in major
bleeding would be statistically signicant indicated that a total of 128
patients would be necessary, assuming 80% power and an = 0.05.
All statistics were performed in Statistical Analysis Software (SAS).
Continuous variables were tested using the t-test or Kruskal-Wallis
test. Categorical variables were tested using X2 or Fishers Exact. Test
selection was based on the validity of the normal assumption. A multivariate logistic regression was performed to assess risk factors potentially associated with major bleeding.
Results

Materials and Methods

Baseline Characteristics

Study Design and Patient Population

Fig. 1 outlines the entry of patients into the study. A total of 710
patients were identied; 289 had an enoxaparin order and 421 had an
UFH order. After exclusion for various reasons (e.g. order not administered, prophylactic dosing, missing order, etc.) 89 enoxaparin treated
patients remained to be matched. A total of 164 patients were included

This single-center retrospective chart review was conducted to evaluate the outcomes associated with use of reduced-dose therapeutic
enoxaparin versus continuous infusion IV UFH for anticoagulation in

T.K. Pon et al. / Thrombosis Research 133 (2014) 10231028

1025

Fig. 1. Flow diagram for patient selection.

in the analysis with 82 patients in each group. Baseline characteristics


were similar between groups with respect to demographics (Table 1).
When comparing baseline comorbidities, coronary artery disease
Table 1
Baseline Characteristics.

Patient Characteristics
Age Mean SD (years)
Height Mean SD (cm)
Weight Mean SD (kg)
Gender n (%)
Comorbidities [n (%)]
Heart Failure
Hypertension
Diabetes
History of CVA
History of ACS
CAD
Liver Disease
Labs (Mean SD)
INR
APTT (seconds)
PLT (k/mm3)
Total Bilirubin
Hgb (g/dL)
Hct (%)
Vital Signs (Mean SD)
Temperature (Celsius)
Heart Rate
Respiratory Rate
Systolic Blood Pressure
Diastolic Blood Pressure

Enoxaparin (n = 82)

UFH (n = 82)

57 16
167.3 11
80.4 21
35 (43)

55 15
167.4 11
81.8 25
35 (44)

0.5
0.98
0.72
1

38 (46.3)
74 (90.2)
51 (62.2)
14 (17.1)
13 (15.7)
34 (41.5)
13 (15.9)

30 (36.6)
64 (78.1)
35 (42.3)
18 (21.9)
9 (10.7)
17 (20.7)
10 (12.2)

0.23
0.05
0.02
0.55
0.37
0.007
0.65

1.49 0.52
57.9 32
227.1 108
0.9 0.57
9.95 1.6
30.2 4.8

1.2 0.31
44.5 29
185.3 71.2
1.6 2.6
10.5 1.86
31.8 5.64

b0.001
0.02
0.005
0.13
0.05
0.06

36.7 0.53
80 14
18 2.4
129.4 26.1
69.8 15.7

36.6 0.63
89.6 22.9
18 3.4
126.5 28.3
68.4 20.9

0.08
0.002
0.99
0.51
0.63

Abbreviations standard deviation (SD), cerebrovascular accident (CVA), acute coronary


syndrome (ACS), coronary artery disease (CAD), international normalized ratio (INR),
activated partial thromboplastin time (APTT), platelet (PLT), hemoglobin (Hgb),
hematocrit (Hct)

(CAD) and diabetes were statistically signicantly more prevalent


in the enoxaparin group. Treatment of VTE was the most common indication for anticoagulation (Fig. 2). The baseline risk for bleeding assessment using the HAS-BLED scoring system showed a similar distribution
in risk for enoxaparin treated and heparin treated patients (Fig. 3) [18].
Renal and Hemodialysis Characteristics
The majority of patients in both groups had documented chronic
kidney disease requiring HD; however, signicantly more patients
treated with UFH were admitted with acute kidney injury requiring
temporary HD (23% vs 7%, p = 0.008) (Table 2). Signicantly more
patients in the enoxaparin group received intermittent hemodialysis
when compared to the UFH arm (95.1% vs 84.2%, p = 0.03). A minority
of patients (9.7%) received other modes of dialysis, which included
peritoneal dialysis (PD), slow-extended daily dialysis (SLEDD), and continuous renal replacement therapy (CRRT).
Enoxaparin and UFH Usage
For the patients receiving enoxaparin, the average number of doses
was 3.3 4.2 per subject, (n = 61, range 123), and the dose ranged
from 0.4 to 1 mg/kg/day, with an average dose of 0.7 0.2 mg/kg/day
(Table 3). The average duration of therapy for UFH was 8.6 8.8 days,
and the target APTT ranges averaged 69 9.2 seconds for the lower
target limit to 89.7 9.1 seconds for the upper target limit.
Outcome Measures
When comparing enoxaparin treated patients to patients treated
with IV UFH, there were no statistically signicant differences in the
incidence of either major bleeding (6.1% vs 11%, p = 0.4) or thromboembolic events (0% vs 2.44%, p = 0.5). With respect to secondary

1026

T.K. Pon et al. / Thrombosis Research 133 (2014) 10231028

Fig. 2. Indications for Anticoagulation. All indications require treatment doses of anticoagulation. Multiple indications frequently included both atrial brillation and VTE treatment. VTE
prophlyaxis refers to post-total knee arthroplasty or post-total hip arthroplasty prophylaxis in which therapeutic anticoagulation levels are targeted.

outcomes, hospital length of stay was signicantly shorter in the


enoxaparin group (20 58.3 vs 28.9 44.5, p = 0.02); there were
no differences in mortality or readmission rates (Table 4).
In the multivariable logistic regression analysis of risk factors for
bleeding, the use of enoxaparin was associated with somewhat lower
risk, but this was not statistically signicant (OR = 0.77, 95%CI:0.23.5, p = 0.73). Antiplatelet use was associated with a signicant
increase in the risk of bleedinging (OR = 18.9, 95%CI 1.8-200, p =
0.03), whereas concomitant anticoagulant (i.e. bridging with parenteral
anticoagulation to warfarin) use was protective (OR = 0.15, 95%CI:
0.03-0.7) (Table 5).
Discussion
The most signicant nding from this retrospective study was that
there was no signicant difference in the incidence of major bleeding
among HD patients treated with therapeutic doses of enoxaparin versus
IV UFH. However, these results should be interpreted cautiously, and
it should be stressed that these chronic HD patients were treated with
a lower adjusted dose of enoxaparin that that averaged 0.7
0.2 mg/kg/day. This dose was comparable to the reported range of
intravenous doses of enoxaparin used for dialysis circuit anticoagulation
in previous studies [7,1116]. In most cases, enoxaparin was ordered as
a parenteral bridging agent for the transition to therapeutic warfarin.
This was often done close to time of hospital discharge, but it was also

commonly done when intravenous access for UFH continuous infusion was lost. Given the fact that most of the patients treated with
enoxaparin had chronic renal failure and were deemed stable for bridging therapy, similar patients are likely the best candidates for treatment
based on the ndings of this study. Our ndings do not support the use
of enoxaparin in critically ill or unstable HD patients, nor do they support the use of enoxaparin in patients receiving CRRT.
Enoxaparin was the selected LMWH based on the larger range of
available syringe dose sizes, and a review of published experiences in
literature that have described safe use of intravenously administered
enoxaparin 0.7 mg/kg given prior to HD in order to prevent thrombosis
of the dialysis circuit at the time [20]. Because randomized clinical trials
exploring use of LMWH at treatment doses uniformly excluded
patients with renal failure who required renal support therapy, and
because the administration of 1 mg/kg once daily in clinical trial
patients who had an estimated CrCl below 30 mL/min was associated
with increased bleeding, the anticoagulation service at UCDMC recommended that enoxaparin be given in the dose range of 0.5 to 1 mg/kg
once daily coupled with clinical assessment of the patients risk for
bleeding and thrombosis [4]. Although the HAS-BLED bleeding risk
score has only been validated to estimate bleeding risk in atrial brillation patients receiving warfarin therapy, it does account for important
risk factors for bleeding [19]. The score was used as an objective
way to estimate bleeding risk for all patients in the study and showed
patients receiving enoxaparin had a similar risk as those receiving
UFH. Patients at high risk for bleeding and/or low risk for thrombosis
received a dose at the lower end of the recommended range whereas
patients with high risk for thrombosis and/or lower bleeding risk
received doses at the higher end.
The dosing range selection used in this study was further supported
by an analysis by Saltissi et al. in which dosing reductions from 1 mg/kg
to 0.69 mg/kg of intravenous enoxaparin administered pre-dialysis

Table 2
Renal insufciency and hemodialysis characteristics.

Fig. 3. Bleeding risk assessment using the HASBLED scoring system. HAS-BLED acronym:
Hypertension (uncontrolled, N160 mmHg systolic), Abnormal renal/liver function, Stroke,
Bleeding history or predisposition (anemia), Labile INR (i.e. therapeutic time in range
b60%), Elderly (N65 years), and Drugs/alcohol concomitantly (e.g. antiplatelet agents,
non-steroidal anti-inammatory drugs). Maximum score = 9.

Type of Renal Failure [n (%)]


Chronic Kidney Disease
Acute Renal Failure
Hemodialysis Mode [n (%)]
Intermittent Hemodialysis (IHD)
Slow Extended Daily Dialysis (SLEDD)
Continuous Renal Replacement Therapy (CRRT)
Peritoneal Dialysis (PD)

Enoxaparin
(n = 82)

UFH
(n = 82)

75 (93)
7 (7)

63 (77)
19 (23)

0.008

78 (95.1)
2 (2.41)
0 (0)
2 (2.41)

69 (84.2)
6 (7.32)
4 (4.88)
3 (3.66)

0.03
0.28
0.12
1

T.K. Pon et al. / Thrombosis Research 133 (2014) 10231028


Table 3
Enoxaparin Usage.

Table 5
Multivariate analysis of bleeding outcome.

Average number of doses (n = 61)


Mean SD
Dose range (mg/kg/day)
Average dose (mg/kg/day)
Mean SD

3.3 4.2
0.4 1
0.7 0.2

were associated with a reduction in minor bleeding and no notable


change in thrombosis rates [13]. Furthermore, Chan et al. observed in
a recent retrospective analysis of patients requiring chronic HD that
use of prophylactic subcutaneous enoxaparin in a dose of 30 mg had a
similar incidence of serious bleeding compared to HD patients treated
with higher doses of 31 to 60 mg, suggesting that the risk of bleeding
is minimally increased when doses of 31-60 mg are used; it should be
noted that this study excluded patients with lower body weight [21].
The use of anti-factor Xa activity levels to adjust dosing was not incorporated into management decisions because of a reported discordance
between anti-Xa levels and other markers of thrombin generation and
a poor correlation with renal function in both HD and non-dialysis
dependent patients with chronic renal insufciency [7,2226]. Studies
have failed to show a consistent relationship between anti-Xa levels
and bleeding [2]. In addition to lack of correlation to a biological effect,
other factors in this population that affect thrombosis or bleeding
may be present, further limiting the value of measuring and adjusting
the dose of LMWH using anti-factor Xa values. [10,20] Monitoring of
enoxaparin in the current study was based only on clinical assessment,
complete blood counts (CBC), and other signs and symptoms of bleeding
or thrombosis.
The study was not sufciently powered to interpret efcacy of
enoxaparin treatment; however, the incidence of major bleeding in
the enoxaparin group was unexpectedly low, and not signicantly
different compared to the UFH group, suggesting an acceptable safety
prole. In the multivariable analysis, the adjusted risk of major bleeding
was slightly but not statistically signicantly reduced in patients treated
with enoxaparin. Cosette and colleagues were able to demonstrate that
use of therapeutic UFH was associated with a higher risk of bleeding in a
univariate analysis [27]. This suggests that further study with a larger
sample size may show a statistically signicant protective effect of
enoxaparin in HD patients. Also, in the multivariable analysis, the observation that transitioning enoxaparin to warfarin was protective against
bleeding appears counterintuitive. This may have occurred because of
the large number of patients in the study were being bridged to warfarin therapy and may have receive anticoagulation for a shorter time
period and have a shorter duration of exposure to heparin or LMWH.
In addition to its retrospective design, this study was subject to other
limitations. Matching was limited to one criterion, anticoagulation indication, in order to achieve an adequate sample size, which led to exclusion of patients treated with UFH. However, without requiring matching
based on the indication for anticoagulation we felt the study would have
been even more limited. The groups were relatively similar with respect

Table 4
Outcome measures.

Primary Endpoints [n (%)]


30-Day ISTH Major Bleeding
30-Day Thromboembolic Event or
Dialysis Catheter Clotting
Secondary Endpoints
Hospital Length of Stay (Days)
Mean SD
30-Day Mortality n (%)
30-Day Readmission n (%)

1027

Treatment (enoxaparin vs UFH)


Age
Female gender
Hgb/Hct
Chronic renal insufciency
History of CVA
Liver disease
Concomitant oral anticoagulant
Antiplatelet therapy

Odds Ratio

95% CI

0.8
0.98
1.6
2.4
12.1
3.2
2.8
0.15
18.9

0.17, 3.5
0.9, 1.04
0.3, 8.1
0.43, 13.1
0.6, 2.3
0.6, 17.5
0.4, 19.2
0.03, 0.74
1.8, 200

0.73
0.59
0.56
0.32
0.09
0.18
0.30
0.02
0.03

to baseline demographics and comorbidities, differing only in a higher


frequency of diabetes and CAD, which put the enoxaparin arm at a
potential increased risk for thrombotic events such as stroke; however,
there were no such complications. Coagulation laboratory parameters
collected at baseline were difcult to interpret as fewer patients in the
enoxaparin (Lovenox) group had complete labs ordered. Ten patients
in the enoxaparin arm did not have baseline INRs documented, and less
than half had an APTT drawn; however, it is logical that APTT was not
collected as it is not an appropriate monitoring parameter for LMWH.
Regardless, coagulation parameters collected actually suggested the
enoxaparin arm was at an increased risk for bleeding.
With respect to HD and renal characteristics, more patients in the
UFH group had acute kidney injury and required continuous renal
replacement. No patients in the enoxaparin group underwent continuous renal replacement as a hemodialysis modality. This may suggest
that patients in the UFH group had a greater severity of illness, which
could have accounted for the longer hospital length of stay. The initial
intention for including length of stay as a secondary outcome was the
thought that for some patients, having enoxaparin as a bridging option
would expedite their discharge. However, given the numerous confounders, no such conclusion can be made. Furthermore, because critically ill patients receiving acute HD and CRRT in this study did not
receive enoxaparin (Lovenox) for therapeutic anticoagulation, its use
should be reserved for more stable HD patients.
Exact duration of enoxaparin therapy was also a limitation of the
study. Again, given the retrospective design, we were only able to evaluate what was documented in the EHR, and if patients were discharged
on enoxaparin without receiving their rst dose in the hospital, we were
unable to capture the exact duration of therapy. As such we were able to
collect this information on only 61 patients in the enoxaparin arm, and
the inpatient exposure days ranged from 1 to 23. Furthermore, we were
only able to assess bleeding in hospitalized patients and in patients who
were readmitted for major bleeding to UCDMC. There is potential that patients could have been treated for major bleeding at an outside hospital;
however, this potential is equal for both the enoxaparin and UFH arms.
This may actually be reective of real world limitations of discharging
patients on medications. Finally, if patients were re-admitted to any outside hospitals for thrombotic or bleeding events after being exposed to
enoxaparin (Lovenox), we were unable to capture those admissions
since the study was limited to a single center.
Conclusion

Enoxaparin
(n = 82)

UFH
(n = 82)

5 (6.1)
0

9 (11)
2 (2.44)

0.4
0.5

20 58.3
5 (6.1)
17 (20.7)

28.9 44.5
12 (14.6)
20 (24.4)

0.02
0.12
0.7

Despite the limitations of a retrospective observational study, this


study is the rst to evaluate the safety of using enoxaparin for therapeutic anticoagulation in HD patients. The ndings suggests that reduced
doses of dose enoxaparin are safe to use in patients on chronic HD
who require therapeutic anticoagulation and not simply a low xed
dose of 30 mg a day to prevent venous thromboembolism. The ndings
also suggest that enoxaparin given at a dose of 0.4-1.0 mg/kg once daily
is a viable option in patients on intermittent HD who are not able to
receive heparin because of the inability to monitor or have IV access
issues. Additionally, use of enoxaparin may be useful as a short term

1028

T.K. Pon et al. / Thrombosis Research 133 (2014) 10231028

home management option in hemodialysis patients who are otherwise


stable and no longer require hospitalization. Further studies are needed
to determine if efcacy of enoxaparin therapy is safe in critically ill
patients who require acute renal replacement therapy, including continuous renal replacement therapy. Finally, the encouraging nding
that use of enoxaparin did not result in a higher incidence of bleeding
compared to regular IV heparin needs to be validated in additional
studies. Antiplatelet therapy should be avoided, when possible, in
hemodialysis patients who require therapeutic anticoagulation with a
heparin preparation.
Conict of Interest Statement
This study was not funded, and the authors have no disclosures.
Acknowledgments
Machelle Wilson, PhD for her assistance with statistical analysis.
References
[1] Kearon C, Akl EA, Comerota AJ, Prandoni P, Bounameaux H, Goldhaber SZ, et al.
Antithrombotic therapy for VTE disease: antithrombotic therapy and prevention of
thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice
guidelines. Chest 2012;141(2 Suppl.):e419S96S.
[2] Garcia DA, Baglin TP, Weitz JI, Samama MM. Parenteral anticoagulants: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest
Physicians evidence-based clinical practice guidelines. Chest 2012;141(2 Suppl.):
e24S43S.
[3] Lyman GH, Khorana AA, Kuderer NM, Lee AY, Arcelus JI, Balaban EP, et al. Venous
thromboembolism prophylaxis and treatment in patients with cancer: American
Society of Clinical Oncology clinical practice guideline update. J Clin Oncol
2013;31:2189204.
[4] Lim W, Dentali F, Eikelboom JW, Crowther MA. Meta-analysis: low-molecularweight heparin and bleeding in patients with severe renal insufciency. Ann Intern
Med 2006;144:67384.
[5] Becker RC, Spencer FA, Gibson M, Rush JE, Sanderink G, Murphy SA, et al. Inuence
of patient characteristics and renal function on factor Xa inhibition pharmacokinetics and pharmacodynamics after enoxaparin administration in non-ST-segment elevation acute coronary syndromes. Am Heart J 2002;143:7539.
[6] Chow SL, Zammit K, West K, Dannenhoffer M, Lopez-Candales A. Correlation of
antifactor Xa concentrations with renal function in patients on enoxaparin. J Clin
Pharmacol 2003;43:58690.
[7] Guillet B, Simon N, Sampol JJ, Lorec-Penet AM, Portugal H, Berland Y, et al. Pharmacokinetics of the low molecular weight heparin enoxaparin during 48 h after bolus
administration as an anticoagulant in haemodialysis. Nephrol Dial Transplant
2003;18:234853.
[8] Rodger MA, Ramsay T, MacKinnon M, Westphal M, Wells PS, McCormick B, et al.
Tinzaparin versus dalteparin for periprocedure prophylaxis of thromboembolic
events in hemodialysis patients: a randomized trial. Am J Kidney Dis
2012;60:42734.
[9] Siguret V, Gouin-Thibault I, Pautas E, Leizorovicz A. No accumulation of the peak
anti-factor Xa activity of tinzaparin in elderly patients with moderate-to-severe
renal impairment: the IRIS substudy. J Thromb Haemost 2011;9:196672.

[10] Davenport A. The rationale for use of low molecular weight heparin for hemodialysis
treatments. Hemodial Int 2013;17:s2832.
[11] Schrader J, Stibbe W, Kandt M, Warneke G, Armstrong V, Muller HJ, et al. Low molecular weight heparin versus standard heparin: a long-term study in hemodialysis and
hemoltration patients. ASAIO Trans 1990;36:2832.
[12] Sagedal S, Hartmann A, Sundstrom K, Bjornsen S, Fauchald P, Brosstad F. A single
dose of dalteparin effectively prevents clotting during hemodialysis. Nephrol Dial
Transplant 1992;14:19437.
[13] Saltissi D, Morgan C, Westhuyzen J, Healy H. Comparison of low-molecular-weight
heparin (enoxaparin sodium) and standard unfractionated heparin for
haemodialysis anticoagulation. Nephrol Dial Transplant 1999;14:2698703.
[14] Davenport A. Review article: low-molecular-weight heparin as an alternative anticoagulant to unfractionated heparin for routine outpatient haemodialysis treatments.
Nephrology 2009;14:45561.
[15] Chuang FR, Chen TC, Lee CH, Ng HY, Wang IK, Chang HW, et al. Clinical safety and
anticoagulation efcacy of low-molecular-weight-heparins in chronic hemodialysis
patients: a single medical center experience. Ren Fail 2011;33:9907.
[16] Sridharan S, Berdeprado J, Sivalingam M, Farrington K. Dalteparin dosing in highux haemodialysis and haemoltration. Nephron Clin Pract 2012;122:537.
[17] Shavit L, Lifschitz M, Lee S. Use of enoxaparin to diminish the incidence of vascular
access stenosis/thrombosis in chronic hemodialysis patients. Int Urol Nephrol
2010;43:499505.
[18] Lip GY, Frison L, Halperin JL, Lane DA. Comparative validation ofa novel risk score for
predicting bleeding risk in anticoagulated patients with atrial brillation: the HASBLED (Hypertension, Abnormal Renal/Liver Function, Stroke, Bleeding History or
Predisposition, Labile INR, Elderly, Drugs/Alcohol Concomitantly) score. J Am Coll
Cardiol 2011;57:17380.
[19] Schulman S, Kearon C. Denition of major bleeding in clinical investigations of
antihemostatic medicinal products in non-surgical patients. J Thromb Haemost
2005;3:6924.
[20] Dager WE, Kiser TH. Systemic anticoagulation considerations in chronic kidney disease. Adv Chronic Kidney Dis 2010;17:4207.
[21] Chan KE, Thadhani RI, Maddux FW. No difference in bleeding risk between subcutaneous enoxaparin and heparin for thromboprophylaxis in end-stage renal disease.
Kidney Int 2013;84:55561.
[22] McMahon LP, Chester K, Walker RG. Effects of different dialysis membranes on
serum concentrations of epoetin alfa, darbepoetin alfa, enoxaparin, and iron sucrose
during dialysis. Am J Kidney Dis 2004;44:50916.
[23] Polkinghorne KR, McMahon LP, Becker GJ. Pharmacokinetic studies of dalteparin
(Fragmin), enoxaparin (Clexane), and danaparoid sodium (Orgaran) in stable
chronic hemodialysis patients. Am J Kidney Dis 2002;40:9905.
[24] Bazinet A, Almanric K, Brunet C, Turcotte I, Martineau J, Caron S, et al. Dosage of
enoxaparin among obese and renal impairment patients. Thromb Res
2005;116:4150.
[25] Klingel R, Schaefer M, Schwarting A, Himmelsbach F, Altes U, Uhlenbusch-Korwer I,
et al. Comparative analysis of procoagulatory activity of haemodilaysis,
haemoltration, and haemodialtration with a polysulfone membrane (APS) and
with different modes of enoxaparin anticoagulation. Nephrol Dial Transplant
2004;19:16470.
[26] Brophy DF, Carr ME, Martin EJ, Venitz J, Gehr TWB. The pharmacokinetics of
enoxaparin do not correlate with its pharmacodynamic effect in patients receiving
dialysis therapies. J Clin Pharmacol 2006;46:88794.
[27] Cosette B, Pelletier ME, Carrier N, Turgeon M, Leclari C, Charron P, et al. Evaluation of
bleeding risk in patients exposed to therapeutic unfractionated or low-molecularweight heparin: a cohort study in the context of a quality improvement initiative.
Ann Pharmacother 2010;44:4941002.

S-ar putea să vă placă și