Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Anaesthesia 2015, 70 (Suppl. 1), 87–95 doi:10.1111/anae.

12898

Review Article
Haemostatic management of cardiac surgical haemorrhage
M. W. Besser,1 E. Ortmann2,3 and A. A. Klein4

1 Consultant, Department of Haematology, 2 Locum Consultant, 4 Consultant, Department of Anaesthesia and Inten-
sive Care, Papworth Hospital, Cambridge, UK
3 Consultant, Department of Anaesthesia and Intensive Care, Kerckhoff Klinik Heart and Lung Centre, Bad Nauheim,
Germany

Summary
Almost 30 000 cardiopulmonary bypass operations are performed in the UK every year, consuming a considerable
portion of the UK blood supply. Each year, in cardiac surgery, 90% of blood products are used by only 10% of
patients, and over the past 25 years, much innovation and research has gone into improving peri-operative diagnosis
and therapy for these patients. Visco-elastic tests performed at the bedside, with modifications to allow direct quanti-
fication of fibrinogen levels, are probably the biggest advancement. There is no clear advantage of thromboelastome-
try over thromboelastography, and the published literature remains scarce. Visco-elastic testing has recently been
coupled with the systematic replacement of clotting factors by means of factor concentrates, with objective improve-
ment in terms of blood loss, red blood cell usage and surgical re-exploration. The National Institute for Health and
Care Excellence has reviewed the available evidence and recommended visco-elastic tests as cost effective in cardiac
surgery. Factor concentrates, however, carry significant risks, particularly unnecessary donor exposures, potential
selective over-correction of partial deficiencies and the possibility that the postoperative risk of venous thrombo-
embolism is increased; as yet there are no data on risk–benefit analysis. There are a number of promising drugs used
in topical haemostasis, but the requirement to apply these before major bleeding is manifest limits their use consider-
ably. Hyperfibrinolysis is less important than in the past due to the wide spread adoption of antifibrinolytic agents
and close intra-operative monitoring of heparin effect.
.................................................................................................................................................................
Correspondence to: M. W. Besser; Email: martin.besser@nhs.net; Accepted: 8 September 2014

Introduction excessive bleeding. Although there is no internationally


Cardiothoracic surgery uses approximately 10% of the agreed management protocol for patients bleeding after
blood supply of the National Blood Service in the UK; cardiac surgery, it has been shown repeatedly in the
90% of these products are consumed by only 10% of last 25 years that algorithm-based transfusion proto-
patients operated on. Over 30 000 cardiac surgical pro- cols are superior to individualised decisions [4]. Cur-
cedures are performed every year in the UK [1, 2]. rently, there is a major trend to replace laboratory-
Transfusion rates vary widely, and seem directly pro- based testing with point-of-care-based tests. Clear evi-
portionate to the expertise of the centre when mea- dence for the benefit of individual products is lacking,
sured by the number of cases per year [3]. A number as well as for the benefits of individual tests in isola-
of blood products and pharmacological agents are tion [5]. A number of topical agents are available
available to minimise coagulopathy associated with which, in combination with intravenous administration

© 2014 The Association of Anaesthetists of Great Britain and Ireland 87


Anaesthesia 2015, 70 (Suppl. 1), 87–95 Besser et al. | Cardiothoracic surgical haemorrhage

of products, are useful to reduce microvascular bleed- [12]. In addition to coagulation changes outlined
ing [6]. This review will give an overview on causes, above, inflammation by complement activation
diagnosis, treatment and current state-of-the-art treat- through contact activation via the alternative pathway,
ment based on available evidence. IL-6 production and white cell activation also takes
place, which causes a prolonged inflammatory
Pathophysiology of the coagulopathy response [9, 13].
of cardiopulmonary bypass
Patients undergoing cardiopulmonary bypass (CPB) Activation of fibrinolysis
are subjected to a number of challenges which are At the same time as activating the clotting cascade,
unique to cardiothoracic surgery. The patient receives a thrombin is also an activator of fibrinolysis. The action
large dose of heparin (300 IU.kg 1), and is haemodi- of thrombin during CPB and the release of by-prod-
luted as soon as they are connected to the bypass cir- ucts of thrombin activity, activate endothelial cells to
cuit. All their blood comes into contact with the plastic release tissue plasminogen activator. This in turn con-
tubing of the bypass circuit, which causes a variable verts plasminogen to plasmin, which acts on fibrin
degree of contact activation. Blood is then propelled to clots that have formed and releases split products such
an oxygenator, where it is trickled through fine capil- as D-dimers and fibrin degradation products. The
laries with secondary flow and a high dwell-time, and activity of tissue plasminogen activator increases by at
past a semi-permeable membrane to allow oxygen least three logarithmic scales in proximity to fibrin,
absorption, and then re-injected into the arterial system helping to minimise the chance of systemic hyperfi-
[7]. Propagation of blood can be with either roller or brinolysis in response to a localised complication.
centrifugal pumps, which vary in their degree of plate- Historically, this mechanism accounted for a sig-
let activation, with the latter increasing the degree of nificant proportion of postoperative bleeding, but it is
platelet activation and risk of clot formation [8]. Typi- less important since the introduction of frequent ACT
cally, a portion of the blood is cooled to 4 °C and monitoring during CPB in the 1970s to prevent under-
mixed with potassium chloride and local anaesthetic to heparinisation. Activation of the fibrinolytic system is
deliver cardioplegia solution to the heart, allowing safe usually self-limiting and rarely extends into the post-
temporary cardiac arrest. The patient emerges at the operative recovery period. Excessive fibrinolysis would
end of CPB with reduced coagulation factor levels but require overcoming the endogenous scavenger mole-
high thrombin levels, which is different to other forms cules that serve to inactivate plasmin, or an over-
of surgery. Despite immediate reversal of heparinisa- whelming release of tissue plasminogen activator, such
tion after CPB, inadvertent re-heparinisation from hep- as in exogenous administration of pharmacological
arin-sequestering storage sites in the patient’s body is doses. Where it gains pathological significance, the
not unusual, due to the high heparin doses involved mechanism is usually two-fold: premature breakdown
[9], especially in obese patients or when unusually large of cross-linked fibrin clots, and impairment of new
doses of heparin were administered during CPB due to fibrin polymerisation due to soluble fibrin and high
heparin resistance [10, 11]. Despite the high heparin fibrin degradation products [14].
levels employed during CPB, platelets are activated,
consumed and show selective loss of alpha granules in Causes of excessive bleeding
response to being exposed to foreign material and the There are three main causes of excessive postoperative
flow conditions in the extracorporeal circuit [9]. bleeding in cardiothoracic surgery:
Chest drain blood losses of up to 400 ml are to be
1 Surgical; half of patients who bleed have a surgical
expected in adults in the first 6 h after surgery. Exces-
cause that can be identified at the time of re-stern-
sive bleeding is usually defined as > 2 ml.kg 1.h 1,
otomy.
and high volume transfusion and/or surgical re-explo-
2 Coagulopathy; patients subjected to prolonged CPB
ration may be required. Excessive bleeding is associ-
time, intra-operative hypothermia, or disseminated
ated with a tripling of mortality, up to 10–15% overall

88 © 2014 The Association of Anaesthetists of Great Britain and Ireland


Besser et al. | Cardiothoracic surgical haemorrhage Anaesthesia 2015, 70 (Suppl. 1), 87–95

intravascular coagulation, may exhibit microvascu- Viscoelastic assays, such as TEG and ROTEM,
lar bleeding in the absence of a surgical cause. have evolved from the simple addition of heparinase
3 Pre-operative undiagnosed or untreated coagulopa- to allow haemostatic assessment during CPB while the
thy; due to anticoagulants, antiplatelet agents, con- patient is fully heparinised. Further refinements of in
genital bleeding diathesis, thrombocytopenia or vitro conditions now produce values corresponding to
reversible platelet dysfunction. the aPTT (INTEM (ROTEM) or kaolin-based TEG);
PT (EXTEM (ROTEM) or rapid-TEG) and fibrinogen
Surgical bleeding can be due to a number of factors,
(FIBTEM (ROTEM) or functional fibrinogen (TEG)).
for example residual chest wall bleeding, bleeding suture
In the past, a laboratory Clauss (functional) fibrinogen
lines or, in extremis, graft dehiscence. If not recognised
result of < 1 g.l 1 was the trigger for treatment, but
in a timely fashion, surgical bleeding can lead to inap-
recent recognition of the importance of fibrinogen has
propriate transfusion and delay in re-sternotomy, with
led to suggested thresholds of 1.5–2.0 g.l 1 [20]. FIB-
protracted high blood loss. Recurrent factors associated
TEM (ROTEM) and functional fibrinogen (TEG)
with increased bleeding risk are underweight patients,
allow the direct assessment of the fibrinogen contribu-
re-do and complex procedures (e.g. multiple valve
tion to clot firmness by blocking platelets through or
replacements or operations involving the aortic arch),
cytochalasin-D (ROTEM) or abciximab (TEG). In one
as well as emergency operations [1, 15, 16].
study, a threshold of 10 mm (FIBTEM) corresponded
Coagulopathy, on the other hand, needs to be
to a Clauss fibrinogen of 2 g.l 1 [21]. Of note, the
corrected to ensure optimal haemostasis as this will
result may depend on the exact platelet inhibitor used
exacerbate surgical bleeding. Abnormal haemostasis
in the assay, and they are therefore not completely
may be diagnosed in the absence of bleeding, and in
interchangeable [22, 23]. Fibrinogen levels can be cor-
this instance, correction will have no benefit to the
rected, either by the use of cryoprecipitate or fibrino-
patient and carry a potential risk of transfusion-trans-
gen concentrates. Some very small studies have
mitted infection, acute lung injury and immune modu-
suggested that aggressive replacement of fibrinogen
lation [5, 17].
with a commercially available concentrate (RiaSTAPTM;
CSL Behring, King of Prussia, PA, USA) may reduce
Diagnosis red cell transfusion and re-sternotomy [24, 25], how-
During chest closure, the experienced surgeon may be ever further studies are required.
able to distinguish between micro-angiopathic and sur- A competing technology is the Sonoclot (Sienco,
gical bleeding. Subjective assessment of the type and Denver, CO, USA), where the viscoelasticity of blood
degree of bleeding, however, may lead to over- or is assessed with a vertically vibrating probe. The
under-treatment. Current standard of care is the inte- Clot-Signature trace looks completely different to the
gration of laboratory-based results into the treatment ROTEM/TEG trace, but produces very similar infor-
algorithm, such as platelet count, fibrinogen, pro- mation. Modifications are also available to allow differ-
thrombin time (PT), activated partial thromboplastin entiated assessment of the coagulation system, and
time (aPTT) and thrombin time. Increasingly, point- particularly platelet retraction [26].
of-care testing is advocated, such as thromboelastome- The National Institute for Health and Care Excel-
try (ROTEMâ; TEM International, Munich, Germany), lence (NICE) has recently reviewed the available evi-
thromboelastography (TEGâ; Haemonetics Corp., dence for use of visco-elastic testing in cardiac surgery
Braintree, MA, USA) or impedance platelet aggregom- and concluded that viscoelastic testing improves the
etry (Multiplateâ; Roche Diagnostics Ltd, Rotkreuz, use of blood products and factor concentrates, reduces
Switzerland) [5, 9, 17, 18]. In an attempt to quantify red cell transfusion, and is cost effective. It also con-
the volume of bleeding when the chest is still open, cluded that the above was true for intra- and postoper-
the chest may be packed with swabs for exactly five ative use, however they did not recommend any
minutes before weighing the swabs and subtracting particular transfusion algorithm. For ROTEM and
their dry weight [19]. TEG, the effect on mortality, length of ICU and hospi-

© 2014 The Association of Anaesthetists of Great Britain and Ireland 89


Anaesthesia 2015, 70 (Suppl. 1), 87–95 Besser et al. | Cardiothoracic surgical haemorrhage

tal stay, and rate of re-sternotomy was not statistically lation factor replacement in major bleeding, and after
significant across all studies [26]. CPB in particular. In the 2004 British Committee for
Near-patient platelet function testing is not yet Standards in Haematology (BCSH) guidelines [35], a
widely adopted in cardiac surgery. Platelet concentrate target of 1 g.l 1 was suggested; in recognition of the
is usually administered if the laboratory platelet count changes in major bleeding management. A new guide-
is < 100 9 10 9, but many clinicians administer plate- line is due to be published, and this is likely to set a
let concentrate whatever the laboratory value, on the trigger for giving fibrinogen (in the form of cryopre-
basis that platelet function is abnormal due to prior cipitate) at 1.5–2.0 g.l 1. The American and Japanese
administration of aspirin or clopidogrel. Point-of-care guidelines state a minimum target of 0.8 g.l 1, and
devices such as Multiplate (Roche Ltd) for assessing German and Austrian/European trauma guidelines
platelet function using are now available. The evidence suggest 2 g.l 1 [36, 37]. Indeed, clot firmness improves
for their benefit is limited [27–29] and thresholds for up until fibrinogen > 3 g.l 1, with clot formation rate
intervention are not yet clearly defined. However, highest at 2 g.l 1 [38]. While there is no evidence of
abnormal results for the stimulation of the adenosine harm, raising the transfusion threshold increases the
diphosphate (ADP) and thrombin receptors (residual number of eligible patients and, given that factor con-
aspirin and clopidogrel activity respectively) may be centrates expose the patient to 20 000 donations or
predictive for higher transfusion requirements [30]. more, this could be potentially hazardous. The manu-
Laboratory assays for hyperfibrinolysis are used in facturers quote the resultant extreme dilution of indi-
clinical trials, but have not yet entered clinical practice. vidual donations as an advantage, but history has
Hyperfibrinolysis may also be apparent on visco-elastic proven that, dependent on the pathogen, this may or
testing, but their sensitivity is low. Both the maximum may not be true. While data on overall survival benefit
lysis in 60 min and the time of onset of lysis have is not available, caution should be exercised, particu-
been used in research studies. When compared with larly given the cost difference between cryoprecipitate
laboratory tests in trauma patients for fibrinolytic acti- and fibrinogen concentrate on a gram-for-gram basis
vation (plasmin/antiplasmin complex and D-dimer lev- and the lack of data on risk-benefit and cost effective-
els), more than 90% had a normal fibrinolytic pattern ness.
in the maximum lysis (ML > 15%) of the ROTEM The choice between cryoprecipitate and fibrinogen
assay [31–33]. concentrate varies from country to country, partially
Cardiothoracic postoperative bleeding may be due to availability. Non-heat-treated fibrinogen con-
complicated by early coagulopathy due to the high centrate was withdrawn in the US and replaced with
intra-operative dose of heparin, whether due to incom- cryoprecipitate to reduce the risk of transmission of
plete reversal with protamine, or inadvertent silent re- non-A, non-B hepatitis, which affected up to 5% of
heparinisation of the patient from sequestration sites. batches in the late 1970s, with a number of countries
Consequently, some of the massive blood loss proto- following suit. Fibrinogen concentrate is now heat-
cols in use by other specialties are of limited benefit in treated, but this has not led to a re-introduction of this
cardiac surgical patients [9]. This means that rapid product in all countries where the license had been
turnaround and appropriate testing is even more previously withdrawn. Cryoprecipitate itself was not
important than in other forms of surgery. originally designed as a treatment for hypofibrinogena-
emia, but was intended as a treatment for haemophilia
Treatment A and von Willebrand disease, and subsequently found
Available clotting factor concentrates, their relative fac- to also be rich in fibrinogen and factor XIII [39].
tor content and cost are compared in Table 1. Interestingly, the NICE guidance on viscoelastic testing
does not refer to cryoprecipitate at all, and recom-
Fibrinogen mends fibrinogen concentrate instead, despite the
Replacing fibrinogen if levels are low has taken centre absence of a UK license for the treatment of acquired
stage with regard to the relative importance of coagu- coagulopathy [26].

90 © 2014 The Association of Anaesthetists of Great Britain and Ireland


Besser et al. | Cardiothoracic surgical haemorrhage Anaesthesia 2015, 70 (Suppl. 1), 87–95

Fresh frozen plasma

Donor exposures

on residual factor content also and will be higher than their standard treated counterparts. Cost estimate based on purchase price does not take reduced content into
Prices are approximate UK list prices and the cost per unit has been estimated based on these. Price estimates are confounded by the wide variation in units from batch to
batch quoted by concentrate manufacturers and the UK National Blood service, and are a guide only. Actual cost per unit/g of methylene blue treated products will depend
There is limited evidence for the therapeutic benefit of

(estimated)
fresh frozen plasma (FFP) in postoperative coagulopa-

> 20 000

> 20 000
thy. The main body of evidence is from retrospective

10
data in massive transfusion, where civilian and military
4

trauma studies showed a survival benefit in patients


£180 (£1080 if born
Cost per bag (FFP),
pool (Cryo), 500 U

who received higher FFP:red cells ratio (so called 1:1


after 01/01/1996-
non-UK-sourced)
blue FFP £178)
£28 (Methylene

ratio) [40–43]. However, this data have been criticised


(Fibrinogen)

in that there was survivorship bias because patients


(PCC) or g

who died on arrival at the emergency centre would


£255

£408

not have had time to receive FFP.


The most frequently administered dose of FFP in
the UK is 15 ml.kg 1 in adults [44, 45]. This is a
non-UK-sourced)
£ (£ if born after
gram fibrinogen

blue FFP: £356)


£56 (Methylene

dose of convenience, which is tolerable in most


01/01/1996-

patients, taking into account that an adult has


Cost per

between 36 and 38 ml.kg 1 plasma volume, and that,


£408

on average, FFP contains 1 U.ml 1 clotting factors.


NA

Prothrombin time and aPTT are screening tests which


born after 01/01/1996-
£0.12 (Methylene blue-

are dependent on the exact reagent–analyser combina-


£0.28 (F.VIII) (£1.71 if
treated FFP: £0.71)

tion, and become prolonged when there is moderate


non-UK-sourced)

reduction of a single clotting factor below the refer-


clotting factor
Cost per unit

ence range. Paradoxically, in a patient with multiple


coagulation factor deficiencies, this occurs at a higher
£0.51

threshold than in single factor deficiencies [46, 47].


NA
Table 1 Relative factor content and cost of current haemostatic treatment.

Therefore, a standard dose of FFP can elevate the


plasma factor concentration to a level where these
1000 ml
Volume

tests return to near normal, but the clinical effect


100
40

50

may be less pronounced. A dose of 30 ml.kg 1 FFP


FFP, fresh frozen plasma; PCC, prothrombin complex concentrate.

has been quoted as potentially a more effective thera-


Fibrinogen

peutic alternative. While this dose makes sense from


content

a coagulation point of view, the major setback is the


2g

2g

1g
NA

large volume of administration (2.25 l for a 75 kg


patient), which could potentially nearly double the
Factor content of therapeutic

circulating plasma volume. This could be detrimental


1000 U (higher if reversing
plasma replacement dose

in patients exhibiting a degree of myocardial stunning


or cardiac failure after cardiac surgery [45]. Given
for 70 kg patient

this, and the fact that CPB causes haemodilution, the


indication for FFP in patients with mild prolongation
warfarin)

of PT, aPTT or visco-elastic parameters is often ques-


1000 U

240 U

tionable.
NA

Prothrombin complex concentrates


Cryoprecipitate

The two most widely used products in Europe are


concentrate

account [34].
Fibrinogen

four-factor prothrombin complex concentrate (PCC)


containing protein C and S, as well as factors VII, IX,
PCC
FFP

X and II and traces of heparin. While PCC can replace

© 2014 The Association of Anaesthetists of Great Britain and Ireland 91


Anaesthesia 2015, 70 (Suppl. 1), 87–95 Besser et al. | Cardiothoracic surgical haemorrhage

the vitamin K-dependent clotting factors regardless of gation is only one of the functions of platelets. Even
the cause of clotting factor depletion, the universally when more than one aggregation response assessment
accepted (and licensed) indication remains warfarin is performed (e.g. collagen, aspirin, ADP, thrombin
reversal. A dose equivalent in vitamin K-dependent receptor agonist peptide), interpretation becomes more
clotting factor activity to 15 ml.kg 1 FFP in a 70 kg and more difficult given the complex nature of post-
patient can be given in 40–50 ml of fluid as opposed CPB coagulopathy [5, 52, 53]. Treatment is still trans-
to over 1 l in the case of FFP. However, there is little fusion of platelet concentrate, whatever the defect, with
data as yet comparing PCC with FFP for the treatment a trend to consider double dose platelets or downward
of acute peri-operative haemorrhage, let alone in the adjustment of the platelet transfusion threshold to
cardiac surgical setting. In addition, published studies compensate for reduced function.
have compared point-of-care with laboratory testing at The best measure of platelet function postopera-
the same time as PCC with FFP, making interpretation tively remains unresolved. Currently, platelet function
of the actual advantages and disadvantages very diffi- testing may be more important for pre-operative risk
cult. Therefore, the main benefit is the speed of stratification. Most measures of platelet function
administration, reliable effect, and small treatment become invalid at platelet counts < 50–100 9 10 9 in
volume, however further research is awaited [48–50]. the test tube [54]. Enriching platelets in vitro to allow
an assessment in this situation introduces significant
Platelets delay, which is why platelet aggregometry, considered
There remains great controversy regarding the platelet the gold standard of platelet function assessment, has
transfusion threshold to be applied in cardiac surgery, never become standard in postoperative patients after
due to the platelet function defect present in all cardiac surgery [55].
patients after CPB [51]. Numeric platelet counts from
the laboratory may not be accurate due to clumping, Antifibrinolytic drugs
particularly if the patient is hypothermic or if the sam- There has been a lot of interest in hyperfibrinolysis
ple was taken during or immediately after CPB, which since the introduction of aprotinin in 1986 [56]. In
can lead to an artifactually low platelet count. The current clinical practice, true hyperfibrinolysis apparent
numerical analysis of platelets in the laboratory does in vitro by TEG is a rare occurrence. It would appear
not take into account a variable degree of platelet dys- that a degree of fibrinolysis is to be expected after car-
function induced by CPB and potentially exacerbated diac surgery, but the definition of systemically appar-
by pre-operative treatment with platelet antagonists ent hyperfibrinolysis is less clear.
and/or uraemia [51]. The monitoring of platelet block- Tranexamic acid (TXA), used almost ubiquitously
ade with impedance aggregometry (Multiplate), platelet in UK cardiac surgical practice, and epsilon amino-
mapping (TEG) or latex agglutination (Verify Nowâ; caproic acid (EACA), are synthetic lysine-analogues
Accumetrics, San Diego, CA, USA), remains outside that reversibly block the lysine binding site of plasmin-
what is considered the standard of care, and the clini- ogen which inhibits the lysis of polymerised fibrin.
cal utility of preserved patient platelet function in vitro They have a plasma half-life of around 2 h, and are
remains to be established. Near-patient platelet excreted in the urine in high concentrations [53]. The
function assessment relies on a minimum number of optimum dose of TXA is unknown despite its wide-
platelets available in the patient for analysis (50– spread international use, with 10 mg.kg 1 bolus fol-
100 9 109.l 1), and can be adversely affected by lowed by 1 mg.kg 1.h 1 as a continuous infusion
extremes of haematocrit when this is performed on being the most commonly used regimen, with little
whole blood. As the platelet count decreases, the evidence for higher doses [57]. Seizures are the main
absence of platelet function in vitro is difficult to inter- reported adverse event with TXA, but thrombotic
pret, whereas preserved platelet function despite events after its use have not been reported. Both agents
thrombocytopenia is considered to confirm normal have been shown to reduce blood loss, when used pro-
platelet function. The problem here also is that aggre- phylactically in cardiac surgery. Tranexamic acid may

92 © 2014 The Association of Anaesthetists of Great Britain and Ireland


Besser et al. | Cardiothoracic surgical haemorrhage Anaesthesia 2015, 70 (Suppl. 1), 87–95

be slightly more effective than EACA in reducing plasminogen activator. Its structure is modified from
blood loss, however there is little evidence to support vasopressin to reduce its vasoactive actions. This reac-
the exceedingly high doses (up to 10 g) used in some tion is exhaustible, and the drug is typically adminis-
centres [57]. tered no more frequently than once in 24 h. It is most
Aprotinin is a bovine-derived serine protease often used in mild haemophilia A, von Willebrand’s dis-
inhibitor which has powerful antiplasmin and antikal- ease and platelet disorders, and it is also a useful adjunct
likrein effects. In high doses, it reduces bleeding in in uraemic and cirrhotic bleeding. Rapid administration
cardiac surgery [57]. It has a number of additional can lead to hypotension, and the antidiuretic hormone-
effects such as reduction in neutrophil activation and like effect can lead to fluid retention [60].
interaction with the renin–angiotensin system. The The evidence for the use of DDAVP in cardiac
half-life of this drug is more than twice as long as surgery comes from two trials which showed that, in
TXA or EACA (5–10 h). Despite a positive effect on small cohorts of patients undergoing complex cardiac
red cell transfusion and reduction in blood loss, a surgery, DDAVP administration led to a reduction in
retrospective analysis of 4000 patients by Mangano mean 24-h blood loss compared with placebo, and
et al. [58] detected an increase in the incidence of those patients with the lowest vWF levels benefitted
renal failure, increased incidence of myocardial infarc- the most [61, 62]. Subsequent trials have failed to
tion and heart failure in the aprotinin group com- show a reduction in blood loss following DDAVP
pared with TXA, EACA and placebo. This was administration. Horrow et al. were able to show that
confirmed in the randomised controlled BART trial co-administration of TXA was necessary in a small
[59], which studied 2331 high-risk cardiac surgery group of patients to reduce blood loss, which the
patients. The trial was terminated early due to excess authors attributed to concomitant tissue plasminogen
mortality in patients who received aprotinin, despite activator release [63].
decreased blood loss and transfusion [59]. This led to
the withdrawal of aprotinin. Recombinant factor VIIa
Currently, aprotinin use in the UK can only be Pharmacological doses of recombinant factor VIIa
considered in uncomplicated coronary artery bypass (rVIIa) concentrate should only be used as a last
graft surgery for named patient use, however this is resort, due to the excess of prothrombotic events and
not ideal because the majority of these cases are not at lack of evidence for a reduction in overall mortality
excessive risk of bleeding. The administration of apro- [64]. Recombinant factor VIIa contains factor levels in
tinin to patients who would probably benefit the most, excess of four logs of the physiological concentration
such as those undergoing surgery for endocarditis, of VIIa, which are able to activate factor Xa in the
re-sternotomy and complex multiple procedures, is not presence of activated platelet surfaces. A recent meta-
currently advocated or licensed. Where aprotinin is analysis included 4468 patients, and showed that rVIIa
administered, care must be taken to assess the effect increased the risk of arterial thrombosis (5.5% vs
on celite-based coagulation tests such as some acti- 3.3%), especially in the elderly, whereas the risk of
vated clotting test cartridges and aPTT reagents, and venous thromboembolism remained unchanged [65].
additional heparin administration may be required. Where rVIIa is used, coagulopathy must be addressed
The prolonged aPTT seen postoperatively, given the first, as there is a greatly reduced effect in the presence
half-life of 10 h, may lead to inappropriate FFP or of hypothermia, hypocalcaemia, hypofibrinogenaemia
protamine administration [57]. or major coagulopathy.

Desmopressin Activated prothrombin complex concentrate


Desmopressin (DDAVP) is a vasopressin analogue Activated prothrombin complex is plasma-derived, and
which releases endogenous von Willebrand Factor contains similar quantities of factor VII, IX and X
(vWF) stored in the Weibel–Pallade bodies of endothe- compared with (non-activated) four-Factor PCC. In
lial cells, as well as factor VIII, prostacyclin and tissue addition, small quantities of factor VIIa and Xa are

© 2014 The Association of Anaesthetists of Great Britain and Ireland 93


Anaesthesia 2015, 70 (Suppl. 1), 87–95 Besser et al. | Cardiothoracic surgical haemorrhage

present. It has been shown to be effective in small Mechanical


cohorts of patients, but also produced thrombosis in Mechanical haemostatic agents (microporous polysac-
unusual sites as a side effect, and its use should be charide, chitin/chitosan-based, collagen, gelatin, cellu-
restricted in a similar way to rVIIa [66]. lose) provide a framework for platelet or red cell
adhesion, with little alteration in haemostasis. Exam-
Factor XIII ples include Surgicel (Johnson & Johnson Medical Inc,
Factor XIII concentration is reduced during CPB, con- Arlington TX, USA) and Oxycel (Oxycel, Pershore,
sistent with other clotting factors. Over the last 20 years, UK).
there have been a few small studies with conflicting
results regarding the link between low factor XIII Flowable
plasma levels and excessive bleeding after cardiac sur- Flowable agents are also based on gelatin from various
gery. In a randomised controlled trial of recombinant animal sources but some, such as Surgifloâ (Ethicon,
factor XIII vs placebo, there was no therapeutic benefit Livingston, UK) or FloSealâ (Baxter, Compton New-
on transfusion avoidance, transfusion requirements or bury, UK), contain plasma-derived human thrombin.
operative revision, despite an observed correction of
factor XIII levels. Factor XIII administration outside Fibrin/albumin/synthetic sealants
clinical trials should therefore be discouraged [67–71]. Fibrin or synthetic sealants are based on the dual
action of fibrinogen and thrombin, and contain human
Topical sealants pooled thrombin and fibrinogen. Examples include
The first use of human plasma as a topical haemostatic Tisseelâ (Baxter) or Evicelâ/Quixilâ (Ethicon). Syn-
agent was documented in 1909 [72]. There are a num- thetic alternatives function by a similar mechanism,
ber of situations considered amenable to topical hae- but do not contain human or animal plasma deriva-
mostatic agents: diffuse raw surface bleeding or oozing; tives. Examples include cyanoacrylate (Omnexâ; Ethi-
venous-type bleeds; bone bleeding; and needle-hole con) and CoSealâ (Proseal, Macclesfield, UK). Tisseel
bleeding. As a rule of thumb, low-flow situations are contains a fibrinolysis inhibitor, bovine aprotinin and
most amenable to topical haemostatic agents, meaning Quixil contains TXA.
that the best time to apply them may be before wean- The sealant Bioglueâ (Cryolife, Kennesaw, GA,
ing from CPB, while there is low flow in the wound USA) contains bovine serum albumin and glut-
bed. However, this is suboptimal, as the unselective araldyde, making it difficult to categorise as it contains
use of these agents is undesirable. Moreover, the sur- albumin rather than fibrin or synthetic sealant but
geon has to weigh risks and benefits carefully before does not rely on the action of added thrombin.
their application, as some of these topical agents may
be inadvertently aspirated by the cell saver or cardioto- Active
my sucker and therefore re-infused to the patient Active topical haemostatic agents are based on bovine
intravascularly. In addition, leakage of these agents (Thrombin JMIâ; Pfizer, Walton Oaks, UK), human
through incompletely sealed wounds into the surgical (Evithromâ; Ethicon) or recombinant thrombin
site and embolisation has been described [73]. (Recothromâ; The Medicines Company, Parsippany,
Topical haemostatic agents rely on the patients’ NJ, USA). A completely synthetic tissue sealant con-
native platelets and haemostatic system to provide a sisting of recombinant fibrinogen, thrombin and factor
framework for platelet or clot adhesion. They are XIII has recently been described [72].
mostly isolated or combined animal, or human, Evidence for the use of these products is limited,
plasma-derived clotting proteins, recombinant clotting and most of the studies have a considerable risk of
proteins or other polymers. Broadly, four types of topi- bias due to sponsorship by manufacturers [6]. Also,
cal agents are in use, even though the nomenclature is the criteria for determining a response are difficult to
somewhat confusing, given the heterogeneity of the standardise, and there are often clinical confounding
components. factors.

94 © 2014 The Association of Anaesthetists of Great Britain and Ireland


Besser et al. | Cardiothoracic surgical haemorrhage Anaesthesia 2015, 70 (Suppl. 1), 87–95

Those products that are based on bovine thrombin reversal of pre-operative anticoagulant therapy by
are associated with a risk of inducing anti-human using agreed protocols.
factor V antibodies due to bovine prothrombin cross-
reacting with human factor V (Thrombin–JMIâ; Conclusion
Pfizer, New York, USA) [74–77]. Since their first The optimal transfusion support of the bleeding
inception in 1943, the purity of the products has patient after cardiothoracic surgery remains to be
improved substantially. The incidence of factor V established. A ‘state-of-the-art’ transfusion algorithm
inhibitor with the currently available bovine thrombin should include indicators for residual heparin, mea-
product (Thrombin-JMI), which contains less bovine sures of haemodilution or coagulation factor consump-
thrombin product, remains unknown [78]. tion, an indicator specific for hypofibrinogenaemia,
Patients who refuse blood products would have and a platelet count. The tests should be rapidly avail-
to be specifically consented with regard to the able and clinicians should be familiar with their inter-
preferred topical sealant, making particular reference pretation and limitations. The routine use of platelet
to the bio-source of the components. The benefit of function tests cannot currently be recommended. More
these agents is likely twofold: reduction in allogeneic evidence, particularly from randomised controlled tri-
blood use, which is not clinically significant in many als, is required to identify the haemostatic agents that
cases as the agents are not applied in low-flow are most appropriate to treat postoperative haemor-
situations; and a reduction in time to chest closure rhage and to identify the optimum postoperative treat-
and subsequent transfer to the intensive care unit ment algorithm, including differentiated point-of-care
[6, 73]. testing. This will allow a more tailored transfusion
strategy, with minimal turnaround times in patients
Application with postoperative haemorrhage following cardiac
Patient blood management is a multidisciplinary, evi- surgery.
dence-based approach to reduce avoidable blood trans-
fusion as much as possible. All hospitals in the UK Acknowledgements
have been asked by NHS Blood and Transplant, the MB has received honoraria from GSK and a travel
UK Department of Health and the UK National Blood Grant from CSL Behring. Papworth Hospital NHS
Transfusion Committee to establish a patient blood Trust has received educational grants from CSL
management programme, to include: patient and Behring and Brightwake Ltd to help fund research
clinician education and intervention; a review of the conducted by EO and AK.
frequency and volume of blood testing; use of near-
patient haemoglobin testing; adherence to approved Competing interests
indications and transfusion triggers when requesting No other external funding or conflicts of interest
blood; optimisation of pre-operative anaemia and declared.

© 2014 The Association of Anaesthetists of Great Britain and Ireland 95


Anaesthesia 2015, 70 (Suppl. 1), e29–e31 Besser et al. | Cardiothoracic surgical haemorrhage

References ing. European Journal of Cardiothoracic Surgery 2011; 39:


1. Despotis G, Avidan M, Eby C. Prediction and management of 924–30.
bleeding in cardiac surgery. Journal of Thrombosis and Hae- 17. Davidson S. State of the art – how I manage coagulopathy in
mostasis 2009; 7(Suppl 1): 111–7. cardiac surgery patients. British Journal of Haematology
2. Hung M, Besser M, Sharples LD, Nair SK, Klein AA. The preva- 2014; 164: 779–89.
lence and association with transfusion, intensive care unit 18. Weber CF, Gorlinger K, Meininger D, et al. Point-of-care test-
stay and mortality of pre-operative anaemia in a cohort of ing: a prospective, randomized clinical trial of efficacy in coag-
cardiac surgery patients. Anaesthesia 2011; 66: 812–8. ulopathic cardiac surgery patients. Anesthesiology 2012; 117:
3. Bennett-Guerrero E, Zhao Y, O’Brien SM, et al. Variation in use 531–47.
of blood transfusion in coronary artery bypass graft surgery. 19. Rahe-Meyer N, Pichlmaier M, Haverich A, et al. Bleeding
Journal of the American Medical Association 2010; 304: management with fibrinogen concentrate targeting a high-
1568–75. normal plasma fibrinogen level: a pilot study. British Journal
4. Shore-Lesserson L, Manspeizer HE, DePerio M, Francis S, Vela- of Anaesthesia 2009; 102: 785–92.
Cantos F, Ergin MA. Thromboelastography-guided transfusion 20. Kozek-Langenecker SA, Afshari A, Albaladejo P, et al. Manage-
algorithm reduces transfusions in complex cardiac surgery. ment of severe perioperative bleeding: guidelines from the
Anesthesia and Analgesia 1999; 88: 312–9. European Society of Anaesthesiology. European Journal of
5. Go€rlinger K, Shore-Lesserson L, Dirkmann D, Hanke AA, Anaesthesiology 2013; 30: 270–382.
Rahe-Meyer N, Tanaka KA. Management of hemorrhage in 21. Solomon C, Rahe-Meyer N, Schochl H, Ranucci M, Gorlinger K.
cardiothoracic surgery. Journal of Cardiothoracic and Vascular Effect of haematocrit on fibrin-based clot firmness in the FIB-
Anesthesia 2013; 27: S20–34. TEM test. Blood Transfusion 2013; 11: 412–8.
6. Barnard J, Millner R. A review of topical hemostatic agents for 22. Schlimp CJ, Solomon C, Ranucci M, Hochleitner G, Redl H,
use in cardiac surgery. The Annals of Thoracic Surgery 2009; Schochl H. The effectiveness of different functional fibrinogen
88: 1377–83. polymerization assays in eliminating platelet contribution to
7. Stammers A, Trowbridge CC. Principles of oxygenator function: clot strength in thromboelastometry. Anesthesia and Analge-
gas exchange, heat transfer and operation. In: Gravlee GP, sia 2014; 118: 269–76.
Davis RF, Stammers AH, Ungerleider RM, eds. Cardiopulmo- 23. Agren A, Wikman AT, Ostlund A, Edgren G. TEG functional
nary Bypass: Principles and Practice. Philadelphia, PA: Wolters fibrinogen analysis may overestimate fibrinogen levels. Anes-
Kluwer, Lippincott Wiliiams & Wilkins, 2008: 47–63. thesia and Analgesia 2014; 118: 933–5.
8. De Somer F, Van Noten G. Blood pumps in cardiopulmonary 24. Gorlinger K, Dirkmann D, Hanke AA, et al. First-line therapy
bypass. In: Gravlee GP, Davis RF, Stammers AH, Ungerleider with coagulation factor concentrates combined with point-of-
RM, eds. Cardiopulmonary Bypass: Principles and Practice. care coagulation testing is associated with decreased alloge-
Philadelphia, PA: Wolters Kluwer, Lippincott Wiliiams & Wil- neic blood transfusion in cardiovascular surgery: a retrospec-
kins, 2008: 35–46. tive, single-center cohort study. Anesthesiology 2011; 115:
9. Besser MW, Klein AA. The coagulopathy of cardiopulmonary 1179–91.
bypass. Critical Reviews in Clinical Laboratory Sciences 2010; 25. Weber CF, Klages M, Zacharowski K. Perioperative coagulation
47: 197–212. management during cardiac surgery. Current Opinion in Ana-
10. Ranucci M, Isgro G, Cazzaniga A, Soro G, Menicanti L, Frigiola esthesiology 2013; 26: 60–4.
A. Predictors for heparin resistance in patients undergoing 26. National Institute for Healthcare and Excellence (NICE). Health
coronary artery bypass grafting. Perfusion 1999; 14: 437–42. Technologies Adoption Programme. NICE Diagnostic Support for
11. Ranucci M, Isgro G, Cazzaniga A, et al. Different patterns of Viscoelastometric Point-of-Care Testing. 2014. http://guidance.
heparin resistance: therapeutic implications. Perfusion 2002; nice.org.uk/htdg13 (accessed 01/09/2014).
17: 199–204. 27. Orlov D, McCluskey SA, Selby R, Yip P, Pendergrast J, Karko-
12. Despotis GJ, Filos KS, Zoys TN, Hogue CW Jr, Spitznagel E, Lap- uti K. Platelet dysfunction as measured by a point-of-care
pas DG. Factors associated with excessive postoperative blood monitor is an independent predictor of high blood loss in
loss and hemostatic transfusion requirements: a multivariate cardiac surgery. Anesthesia and Analgesia 2014; 118: 257–
analysis in cardiac surgical patients. Anesthesia and Analgesia 63.
1996; 82: 13–21. 28. Romlin BS, Soderlund F, Wahlander H, Nilsson B, Baghaei F, Je-
13. Holzheimer RG, Go €rlach H, Wilkert S, Hehrlein F, Molloy RG. ppsson A. Platelet count and function in paediatric cardiac
RG IL-6 and TNFa release in association with neutrophil acti- surgery: a prospective observational study. British Journal of
vation after cardiopulmonary bypass surgery. Infection 1994; Anaesthesia 2014; pii: aeu194. [Epub ahead of print]
22: 37–42. 29. Kong R, Trimmings A, Hutchinson N, et al. Consensus recom-
14. Horrow JC, Fitch JCK. Management of coagulopathy associated mendations for using the Multiplate for platelet function
with cardiopulmonary bypass. In: Gravlee GP, Davis RF, Kurusz monitoring before cardiac surgery. International Journal of
CCP, Utley JR, eds. Cardiopulmonary Bypass: Principles and Laboratory Hematology 2014 July 23; doi: 10.1111/ijlh.
Practice. Philadelphia, PA: Lippincott, Williams & Wilkins, 12279. [Epub ahead of print].
2000: 459–72. 30. Schimmer C, Hamouda K, Sommer SP, Ozkur M, Hain J, Leyh
15. Karkouti K, Wijeysundera DN, Yau TM, McCluskey SA, van R. The predictive value of multiple electrode platelet aggre-
Rensburg A, Beattie WS. The influence of baseline hemoglo- gometry (Multiplate) in adult cardiac surgery. The Thoracic
bin concentration on tolerance of anemia in cardiac surgery. and Cardiovascular Surgeon 2013; 61: 733–43.
Transfusion 2008; 48: 666–72. 31. Dekker SE, Viersen VA, Duvekot A, et al. Lysis onset time as
16. Vuylsteke A, Pagel C, Gerrard C, et al. The Papworth Bleeding diagnostic rotational thromboelastometry parameter for fast
Risk Score: a stratification scheme for identifying cardiac sur- detection of hyperfibrinolysis. Anesthesiology 2014; 121: 89–
gery patients at risk of excessive early postoperative bleed- 97.

e29 © 2014 The Association of Anaesthetists of Great Britain and Ireland


Besser et al. | Cardiothoracic surgical haemorrhage Anaesthesia 2015, 70 (Suppl. 1), e29–e31

32. Rijken DC, Lijnen HR. New insights into the molecular mecha- 51. Ortmann E, Klein AA, Sharples LD, et al. Point-of-care assess-
nisms of the fibrinolytic system. Journal of Thrombosis and ment of hypothermia and protamine-induced platelet dys-
Haemostasis 2009; 7: 4–13. function with multiple electrode aggregometry (Multiplate(R))
33. Raza I, Davenport R, Rourke C, et al. The incidence and mag- in patients undergoing cardiopulmonary bypass. Anesthesia
nitude of fibrinolytic activation in trauma patients. Journal of and Analgesia 2013; 116: 533–40.
Thrombosis and Haemostasis 2013; 11: 307–14. 52. Solomon C, Hartmann J, Osthaus A, et al. Platelet concen-
34. Hornsey P, Krailadsiri S, MacDonald J, et al. Coagulation factor trates transfusion in cardiac surgery in relation to preopera-
content of cryoprecipitate prepared from methylene blue plus tive point-of-care assessment of platelet adhesion and
light virus-inactivated plasma. British Journal of Haematology aggregation. Platelets 2010; 21: 221–8.
2000; 109: 665–70. 53. Ranucci M, Aronson S, Dietrich W, et al. Patient blood man-
35. O’Shaughnessy DF, Atterbury C, Bolton Maggs P, et al. Guide- agement during cardiac surgery: do we have enough evi-
lines for the use of fresh-frozen plasma, cryoprecipitate and dence for clinical practice? Journal of Thoracic and
cryosupernatant. British Journal of Haematology 2004; 126: Cardiovascular Surgery 2011; 142: e1–32.
11–28. 54. Hanke AA, Roberg K, Monaca E, et al. Impact of platelet
36. Levy JH, Szlam F, Tanaka KA, Sniecienski RM. Fibrinogen and count on results obtained from multiple electrode platelet
hemostasis: a primary hemostatic target for the management aggregometry (Multiplate). European Journal of Medical
of acquired bleeding. Anesthesia and Analgesia 2012; 114: Research 2010; 15: 214–9.
261–74. 55. Reece MJ, Klein AA, Salviz EA, et al. Near-patient platelet
37. Schlimp CJ, Schochl H. The role of fibrinogen in trauma- function testing in patients undergoing coronary artery sur-
induced coagulopathy. Hamostaseologie 2014; 34: 29–39. gery: a pilot study. Anaesthesia 2011; 66: 97–103.
38. Solomon C, Cadamuro J, Ziegler B, et al. A comparison of 56. Gans H, Krivit W. Problems in hemostasis during openheart sur-
fibrinogen measurement methods with fibrin clot elasticity gery. III. Epsilon amino caproic acid as an inhibitor of plasmino-
assessed by thromboelastometry, before and after adminis- gen activator activity. Annals of Surgery 1962; 155: 268–76.
tration of fibrinogen concentrate in cardiac surgery patients. 57. Ortmann E, Besser MW, Klein AA. Antifibrinolytic agents in
Transfusion 2011; 51: 1695–706. current anaesthetic practice. British Journal of Anaesthesia
39. Sorensen B, Bevan D. A critical evaluation of cryoprecipitate 2013; 111: 549–63.
for replacement of fibrinogen. British Journal of Haematology 58. Mangano DT, Tudor IC, Dietzel C; Multicenter Study of Perioper-
2010; 149: 834–843. ative Ischemia Research Group, Ischemia Reseacrh and Educa-
40. Miller RD. The impact of Vietnam military data on modern tion Foundation. The risk associated with aprotinin in cardiac
civilian transfusion medicine. Anesthesiology 2009; 110: surgery. New England Journal of Medicine 2006; 354: 353–65.
1412–6. 59. Fergusson DA, Hebert PC, Mazer CD, et al. A comparison of
41. Lal DS, Shaz BH. Massive transfusion: blood component ratios. aprotinin and lysine analogues in high-risk cardiac surgery.
Current Opinion in Hematology 2013; 20: 521–5. New England Journal of Medicine 2008; 358: 2319–31.
42. Wang WT, Zhan YH, Hu XB, et al. Fixed rate of blood 60. Ogawa S, Ohnishi T, Hosokawa K, Szlam F, Chen EP, Tanaka
component improves the survival rate of patients in mas- KA. Haemodilution-induced changes in coagulation and
sive transfusion. Biomedical Sciences Reports 2013; 1: 243–6. effects of haemostatic components under flow conditions.
43. Baumann Kreuziger LM, Morton CT, Subramanian AT, Anderson British Journal of Anaesthesia 2013; 111: 1013–23.
CP, Dries DJ. Not only in trauma patients: hospital-wide imple- 61. Salzman EW, Weinstein MJ, Weintraub RM, et al. Treatment
mentation of a massive transfusion protocol. Transfusion with desmopressin acetate to reduce blood loss after cardiac
Medicine 2014; 24: 162–8. surgery. New England Journal of Medicine 1986; 314: 1402–
44. Chowdary P, Saayman AG, Paulus U, Findlay GP, Collins PW. 6.
Efficacy of standard dose and 30 ml/kg fresh frozen plasma 62. Harker LA. Bleeding after cardiopulmonary bypass. New Eng-
in correcting laboratory parameters of haemostasis in critically land Journal of Medicine 1986; 314: 1446–8.
ill patients. British Journal of Haematology 2004; 125: 69–73. 63. Horrow JC, Van Riper DF, Strong MD, Brodsky I, Parmet JL. He-
45. Stanworth SJ, Walsh TS, Prescott RJ, et al. A national study of mostatic effects of tranexamic acid and desmopressin during
plasma use in critical care: clinical indications, dose and effect cardiac surgery. Circulation 1991; 84: 2063–70.
on prothrombin time. Critical Care 2011; 15: R108. 64. Gill R, Herbertson M, Vuylsteke A, et al. Safety and efficacy of
46. Burns ER, Goldberg SN, Wenz B. Paradoxic effect of multiple recombinant activated factor VII: a randomized placebo-con-
mild coagulation factor deficiencies on the prothrombin time trolled trial in the setting of bleeding after cardiac surgery.
and activated partial thromboplastin time. American Journal Circulation 2009; 120: 21–7.
of Clinical Pathology 1993; 100: 94–8. 65. Levi M, Levy JH, Andersen HF, Truloff D. Safety of recombinant
47. Thorell SE, Nash MJ, Thachil J. Clinical implications of clotting activated factor VII in randomized clinical trials. New England
screens. International Journal of Laboratory Hematology 2014 Journal of Medicine 2010; 363: 1791–800.
March 17; doi 10.1111/ijlh.12206. 66. Negrier C, Dargaud Y, Bordet JC. Basic aspects of bypassing
48. Bruce D, Nokes TJ. Prothrombin complex concentrate (Beriplex agents. Haemophilia 2006; 12(Suppl 6): 48–52.
P/N) in severe bleeding: experience in a large tertiary hospi- 67. Grossmann E, Akyol D, Eder L, et al. Thromboelastometric
tal. Critical Care 2008; 12: R105. detection of clotting factor XIII deficiency in cardiac surgery
49. Lin DM, Murphy LS, Tran MH. Use of prothrombin complex patients. Transfusion Medicine 2013; 23: 407–15.
concentrates and fibrinogen concentrates in the perioperative 68. Karkouti K, von Heymann C, Jespersen CM, et al. Efficacy and
setting: a systematic review. Transfusion Medicine Reviews safety of recombinant factor XIII on reducing blood transfu-
2013; 27: 91–104. sions in cardiac surgery: a randomized, placebo-controlled,
50. Makris M. Optimisation of the prothrombin complex concen- multicenter clinical trial. Journal of Thoracic and Cardiovascu-
trate dose for warfarin reversal. Thrombosis Research 2005; lar Surgery 2013; 146: 927–39.
115: 451–3.

© 2014 The Association of Anaesthetists of Great Britain and Ireland e30


Anaesthesia 2015, 70 (Suppl. 1), e29–e31 Besser et al. | Cardiothoracic surgical haemorrhage

69. Levy JH, Gill R, Nussmeier NA, et al. Repletion of factor XIII 74. Banninger H, Hardegger T, Tobler A, et al. Fibrin glue in sur-
following cardiopulmonary bypass using a recombinant A-sub- gery: frequent development of inhibitors of bovine thrombin
unit homodimer. A preliminary report. Thrombosis and Hae- and human factor V. British Journal of Haematology 1993; 85:
mostasis 2009; 102: 765–71. 528–32.
70. Godje O, Gallmeier U, Schelian M, Grunewald M, Mair H. Coag- 75. Knobl P, Lechner K. Acquired factor V inhibitors. Bailliere’s
ulation factor XIII reduces postoperative bleeding after coro- Clinical Haematology 1998; 11: 305–18.
nary surgery with extracorporeal circulation. Thoracic and 76. Lawson JH, Lynn KA, Vanmatre RM, et al. Antihuman factor V
Cardiovascular Surgery 2006; 54: 26–33. antibodies after use of relatively pure bovine thrombin.
71. Shainoff JR, Estafanous FG, Yared JP, DiBello PM, Kottke-Mar- Annals of Thoracic Surgery 2005; 79: 1037–8.
chant K, Loop FD. Low factor XIIIA levels are associated with 77. Dorion RP, Hamati HF, Landis B, Frey C, Heydt D, Carey D. Risk
increased blood loss after coronary artery bypass grafting. and clinical significance of developing antibodies induced by
Journal of Thoracic and Cardiovascular Surgery 1994; 108: topical thrombin preparations. Archives of Pathology and
437–45. Laboratory Medicine 1998; 122: 887–94.
72. Carlson MA, Calcaterra J, Johanning JM, Pipinos II, Cordes CM, 78. Plesca D. A review of topical thrombin. Cleveland Clinic
Velander WH. A totally recombinant human fibrin sealant. Pharmacotherapy Update 2009; XII: 1–4.
Journal of Surgery Research 2014; 187: 334–42.
73. Forcillo J, Perrault LP. Armentarium of topical hemostatic prod-
ucts in cardiovascular surgery: an update. Transfusion and
Apheresis Science 2014; 50: 26–31.

e31 © 2014 The Association of Anaesthetists of Great Britain and Ireland

You might also like