Vous êtes sur la page 1sur 31

 

 
Haematological effects and complications of snake envenoming

I. Berling, G.K. Isbister

PII: S0887-7963(14)00097-2
DOI: doi: 10.1016/j.tmrv.2014.09.005
Reference: YTMRV 50420

To appear in: Transfusion Medicine Reviews

Please cite this article as: Berling I, Isbister GK, Haematological effects and
complications of snake envenoming, Transfusion Medicine Reviews (2014), doi:
10.1016/j.tmrv.2014.09.005

This is a PDF file of an unedited manuscript that has been accepted for publication.
As a service to our customers we are providing this early version of the manuscript.
The manuscript will undergo copyediting, typesetting, and review of the resulting proof
before it is published in its final form. Please note that during the production process
errors may be discovered which could affect the content, and all legal disclaimers that
apply to the journal pertain.
ACCEPTED MANUSCRIPT

Haematological effects and complications of snake envenoming

Berling I1, 2, Isbister GK1, 2

T
1
Department of Clinical Toxicology and Pharmacology, Calvary Mater Newcastle, Newcastle, NSW,

P
Australia

RI
2
School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia

SC
NU
MA
ED
PT
CE
AC

Corresponding author:

Dr Ingrid Berling

Department of Clinical Toxicology, Calvary Mater Newcastle, Edith St. Waratah,

NSW 2298, Australia

Tel.: + 612 49211211

Fax: + 612 49602088

E-mail: Ingrid.berling@gmail.com
ACCEPTED MANUSCRIPT

Abstract

Haematological abnormalities are the most common effects of snake envenoming globally.

Venom induced consumption coagulopathy (VICC) is the commonest and most important.

T
Other haematological abnormalities are an anticoagulant coagulopathy and thrombotic

P
RI
microangiopathy. VICC is venom induced activation of the clotting pathway by procoagulant

toxins, resulting in clotting factor consumption and coagulopathy. The type of procoagulant

SC
toxin differs between snakes and can activate prothrombin, factor X, factor V or consume

NU
fibrinogen. The most useful investigation in VICC is a prothrombin time/international

normalised ratio (INR). The D-Dimer may assist in early diagnosis but fibrinogen levels often
MA
add little in the clinical setting. Bedside investigations would be ideal but point of care testing

INR and whole blood clotting tests (WBCT) have been shown to be unreliable in VICC. The
ED

major complication of VICC is haemorrhage, including intracranial haemorrhage which is

often fatal. The role of antivenom in VICC is controversial and may only be beneficial for
PT

some types of snakes including Echis spp. where the duration of abnormal clotting is reduced
CE

from over a week to 24-48 hours. In contrast, antivenom does not appear to speed the

recovery of VICC in Australian snake envenoming. Other treatments for VICC include factor
AC

replacement, observation and prevention of trauma, and heparin. An Australian study showed

that fresh frozen plasma speeds recovery of VICC but early use may increase consumption.

There is no evidence to support heparin.

Keywords: Snake, venom, coagulopathy, envenoming.


ACCEPTED MANUSCRIPT

Introduction

Globally snakebite results in a significant burden of illness, particularly in tropical and

subtropical regions such as South Asia, South-East Asia and Sub-Sahara Africa [1]. It is

T
estimated that annually there are more than 400,000 snake envenomings and 20,000 deaths

P
RI
worldwide [1]. One of the most common and clinically significant complications of snake

envenoming is coagulopathy, which can result in life-threatening haemorrhage and death [2].

SC
Venom-induced consumption coagulopathy (VICC) is the most frequent type of

NU
coagulopathy associated with snake envenoming globally, but less clinically significant

coagulopathies are also recognised with certain snakes, such as the Australian black snakes
MA
[3, 4]. More recently there has been a recognition of a thrombotic microangiopathic

syndrome occurring in some patients who develop VICC [5].


ED

A recognition and understanding of the pathophysiology of VICC is important for treating


PT

patients envenomed from a snake bite. This review will focus on describing the underlying

mechanisms of coagulopathy in snake bite, how this differs between snake species, and the
CE

investigation and treatment of coagulopathy and associated haematological complications.


AC

Medically important snakes

Snake is the common term used for the ‘serpentes’ family of reptiles. Only about 20% of

snakes are venomous and therefore medically important. Venomous snakes have specialised

venom producing glands and teeth (fangs) for effective venom delivery. Venomous snakes

come from the families Elapidae, Viperidae, Colubridae and Atractaspididae, although most

of the burden of snake envenoming is due to bites by vipers and elapids. As a general rule

vipers mainly cause coagulopathy compared to elapids that mainly cause neurotoxicity

(excluding Australian elapids).


ACCEPTED MANUSCRIPT

Venom-Induced Consumption Coagulopathy

VICC is the broad, simple descriptive term used to describe any coagulopathy that results

from the consumption of clotting factors due to a procoagulant toxin in a snake venom. It is

T
caused by the bites of a range of snakes including Viperid snakes, Elapidae snakes and some

P
RI
Colubrid snakes[2]. Previously the coagulopathy associated with snake envenoming was

often referred to as a disseminated intravascular coagulation (DIC). However the term DIC is

SC
not appropriate for snakebite coagulopathy [5]. DIC results from widespread immune,

NU
endothelial, cellular and clotting cascade activation and has a very high mortality, whereas

VICC has a completely different pathogenesis (specific enzyme activation), is treated


MA
differently and has a much lower mortality..

VICC is characterised by the activation of the clotting pathway due to procoagulant toxins in
ED

the snake venom. They are referred to as procoagulant toxins because in vitro they result in
PT

rapid clot formation, but in vivo lead to severe factor consumption and therefore a risk of

bleeding. The toxins differ between snake species and activate various parts of the clotting
CE

pathway by targeting different clotting factors. Despite these differences, all procoagulant
AC

toxins cause a similar clinical picture of clotting factor consumption and coagulopathy. The

clinical severity of VICC is dependent on which clotting factors are consumed. For example a

milder VICC appears to occur when there is only fibrinogen consumption, whereas a more

severe form of VICC is usually associated with multiple factor consumption, such as is seen

with Australian elapid envenoming which results in FII, FV, FVIII and fibrinogen

consumption, or in Russell’s viper envenoming with FV, FVIII, FX and fibrinogen

consumption.

The toxins in snake venom that produce VICC are classically categorised by where they act

on the clotting pathway, with the most important ones being prothrombin activators, factor V
ACCEPTED MANUSCRIPT

and X activators and thrombin-like enzymes (TLEs), the latter also referred to as

fibrinogenases (Figure 1) [6].

Prothrombin Activators

P T
Two large important groups of snakes contain prothrombin activators: most of the

RI
Australasian elapids (brown snake [Pseudechis spp.], tiger snakes [Notechis spp.], rough-

SC
scaled snake [Tropidechis carinatus], broad-headed snakes [Hoplocephalus spp.] and taipans

[Oxyuranus spp.]) and the Echis genus which includes carpet vipers and the saw-scaled viper

NU
[6]. Prothrombin activators can be classified into four groups (A-D) depending on their

structure, function and co-factor requirements [6]. Australian snakes contain the group C and
MA
D prothrombin activators which are serine proteases. Group C prothrombin activators are

found in the brown snake and taipan and resemble the human prothrombinase or factor XaVa
ED

complex which converts prothrombin to thrombin. Group D toxins are found in tiger snake,
PT

rough-scale snake and broad-headed snakes and contain a toxin similar to human factor FXa,

without the FVa cofactor [7]. Group A and B toxins are found in the Echis species of snakes
CE

and directly activated prothrombin but convert it to meizothrombin rather than the fully
AC

active thrombin [8].

Activation of the clotting cascade by prothrombin activators results in multiple factor

deficiencies due to the positive and negative feedback loops activated when prothrombin (II)

is converted to thrombin (IIa) (see Figure 1) [7]. This leads to activation of FV to FVa and

FVIII to FVIIIa regardless of whether the initial toxin contained a FVa-like complex.

Thrombin also activates fibrinogen to fibrin consuming all of the fibrinogen. Measurements

of factor levels post envenoming have shown severe deficiencies of factor V and factor VIII,

with an associated unrecordable prothrombin time/international normalised ratio (INR) and

undetectable fibrinogen levels, consistent with complete VICC [7]. A partial VICC has been
ACCEPTED MANUSCRIPT

recognised in patients with low, but recordable fibrinogen levels where the pathway has not

been completely activated and not all of the fibrinogen has been converted to fibrin [7, 9].

Factor X and V activators

P T
Russell’s viper (Daboia genus) venom contains factor V and X activators [6]. The more

RI
important toxin is the factor X activator which converts factor X to Xa resulting in the

SC
formation of the prothrombinase complex and subsequent conversion of fibrinogen to fibrin

(see Figure 1). Because the factor X activator toxin acts so earlier in the coagulation pathway

NU
(before the common pathway), it results in positive and negative feedback loops leading to

indirect consumption of factors V and VIII. There is also direct activation and consumption
MA
of factor V. This again results in an unrecordable prothrombin time/INR and undetectable

fibrinogen.
ED

Thrombin-like Enzymes
PT

Thrombin-like enzymes (TLEs) are different to the prothrombin activators and factor Xa/Va
CE

activator toxins because they simply consume fibrinogen rather than activating the clotting

pathway. For this reason, TLEs only produce an isolated deficiency of fibrinogen and
AC

uncommonly affect other clotting factors. They usually cause a milder form of VICC, but

they will result in an unrecordable prothrombin time/INR and bleeding complications if there

is undetectable fibrinogen [10]. There are over 67 different TLE toxins that have been

identified, the majority being zinc metalloproteinase that cleave the alpha chain of fibrinogen

resulting in consumption of fibrinogen without the production of (or conversion to) fibrin

[11]. Other TLEs cleave the beta chain and a few cleave both chains, but they do not result in

intact fibrin. Ancrod is the best known of the TLEs and was for a while tested for use in

cerebrovascular disease until it was shown that there were too many bleeding complications

[12]. Table 1 groups each snake by its toxin type.


ACCEPTED MANUSCRIPT

Clinical Manifestations of VICC

The clinical outcome for patients who develop VICC is not only dependent on the severity of

the coagulopathy, but other factors such as trauma, platelet count and function, and vascular

T
injury are important. Arguably the most important issue in patients with VICC is whether

P
RI
physical trauma occurs and anything that potentially causes vessel damage and the release of

tissue factor may result in haemorrhage. This may result in significant morbidity or death in

SC
the case of intracranial or major organ haemorrhage. Table 2 lists the clinical effects of snake

NU
bite associated with coagulopathy and bleeding. Bleeding can range from minor, such as

oozing at the bite site or cannula insertion site, to major life threatening haemorrhage
MA
requiring blood transfusion. Intracranial haemorrhage can be seen after an associated head

injury, but spontaneous bleeds also occur, often in the setting of hypertension. Thoracic
ED

cavity bleeding or bleeding into other body compartments can occur after trauma.
PT

The metalloproteinase prothrombin activator toxin ecarin found in Echis species not only

activates the clotting pathway, but also acts as a haemorrhagin [7]. These haemorrhagins are
CE

proteolytic enzymes that cause damage to the blood vessel wall, affecting the basement

membrane and creating a ‘shear-stress’ like injury which in turn increases the risk of bleeding
AC

associated with VICC. This explains why spontaneous and more severe bleeding is reported

with Echis envenoming compared to Australian elapid envenoming, despite the prothrombin

activators being more potent in the latter [13, 14].

The duration of VICC depends on the type of toxin and the administration of antivenom.

VICC resulting from Australian elapid envenoming appears to resolve over 24 to 48 hours

independent of antivenom treatment, and the time to recovery appears to depend only on the

production of new clotting factors [8]. On the other hand, Echis induced VICC has been

shown to continue for days if not treated with antivenom [4]. However, after Echis
ACCEPTED MANUSCRIPT

envenoming is treated with antivenom the time of recovery is also 24 to 48 hours

demonstrating that once the toxin is neutralised there is normal recovery of clotting factors.

An understanding of the expected natural course of VICC determined by the type of snake

T
and procoagulant toxin may influence the need for antivenom, duration of hazard reduction

P
for trauma and decision to use factor replacement.

RI
Investigation of VICC

SC
The diagnosis of VICC can be made based on a history of snakebite and evidence of

NU
coagulopathy due to factor consumption. This can be confirmed by an abnormal (often

unrecordable) INR, or prothrombin time (PT), low or undetectable fibrinogen and an elevated
MA
D-dimer (at least 10 times the upper limit of normal) [7]. This definition captures all forms of

VICC, from the milder TLE induced fibrinogen consumption to the more severe multi-factor
ED

consumption, seen with prothrombin activator venoms. Fibrinogen is the most consistently
PT

consumed factor in all types of snake bites causing VICC because it is the common point of

the clotting pathway.


CE

The D-dimer is a marker of fibrin degradation from normally formed cross-linked fibrin clot.
AC

Therefore, D-dimer levels are expected to be markedly increased in VICC resulting from

procoagulant toxins that activate the clotting pathway high up. In this case normal fibrin is

formed from fibrinogen, cross-linked by factor XIII and degraded by plasmin. In the case of

Australian elapids the D-dimer is 100 to 1000 times the upper limit of normal [7]. For

Russell’s viper envenoming the D-dimer is elevated by 10 to 100 times the upper limit. For

the TLEs which consume fibrinogen without the production of fibrin, there is no formation of

cross-linked fibrin and so there is only a modest elevation of the D-dimer [10, 15]. The

reason for an abnormal D-dimer with TLEs is unclear but may be due to the D-dimer assay

being less specific with large concentrations of non-cross linked fibrinogen degradation
ACCEPTED MANUSCRIPT

products or to some associated endogenous activation of the clotting pathway. A less specific

marker of fibrin(ogen) degradation products, FnDP/FgD or FDP assay, will be 100 times

normal in VICC due to TLEs, compared to only a modest increase in the D-dimer [10].

T
However, these FDP assays are no longer available in diagnostic laboratories which is

P
unfortunate for regions where snakes with TLEs occur.

RI
Serial measurement of fibrinogen levels has been used in many studies to demonstrate

SC
recovery of the coagulopathy and is a useful outcome for research into VICC [13, 16].

Clinically, the measurement of fibrinogen in VICC will confirm that there has been

NU
consumption but will not usually add much additional information to a PT/INR or D-dimer.
MA
Fibrinogen is the slowest of all factors to recover and effective clotting appears to occur with

only minimal fibrinogen recovery to 0.5 to 1.0g/L when the PT/INR has almost normalised
ED

[7]. This means that fibrinogen is not a good clinical marker of recovery. The PT/INR is a

more useful marker of recovery of VICC because it measures the functional recovery of the
PT

clotting pathway and clinicians better understand the risk of bleeding associated with elevated

INRs.
CE

Bedside assessment of clotting function in VICC is ultimately the best investigation, but there
AC

continues to be no reliable and accurate bedside test for the diagnosis of VICC. The whole

blood clotting test (WBCT) is the most commonly used bedside clotting test worldwide. This

test works on the principle that blood taken from a patient with VICC is incoagulable and so

will not clot in a glass tube within a defined time frame, usually 20 minutes. Although widely

used, there is no standardised procedure for the WBCT and hence the test is fraught with

error, misinterpretation and high false negative rates in a busy clinical environment, which is

prone to interruption and distraction. Various studies have used time end-points of 10, 15, 20

and 30 minutes and there is no universally agreed tube for collection or defined volume of

blood to be collected. A recent study of the 20 minute WBCT in patients envenomed by


ACCEPTED MANUSCRIPT

Russell’s vipers showed a sensitivity of only 40%, which contributed to delays in antivenom

administration [17]. However, it has been suggested that when performed under strict

procedural guidelines the WBCT may be more accurate and further studies are required to

T
confirm this.

P
RI
Modern point of care (POC) INR testing devices have also been used in patients with VICC.

Unfortunately POC INR machines have also been unreliable and have been shown to have a

SC
high false negative rate in Australian snakebites and cannot be recommended for use. In a

NU
small study of 15 patients the POC INR was normal in 3 of 7 patients with VICC where a

laboratory INR was unrecordable [18]. The reason for this lies in the difference between the
MA
assays from formal laboratories and POC machines (see figure 2). A standard laboratory

prothrombin time is measured by adding thromboplastin (tissue factor) to plasma and


ED

calculating the time to clot formation, usually identifying a clot by either spectrophotometry

(visual absorbance) or fibrometery (mechanical method) [14]. Hence, in a patient with VICC
PT

there is incoagulable blood and no clot is formed.


CE

In contrast, POC INR machines do not assesses or measure clot formation, but use thrombin
AC

cleavage as a marker of clotting function. This is achieved by adding tissue factor to a blood

sample (similar to the laboratory method) to trigger clot formation, and then adding an

electrochemical substrate which is activated when it is cleaved by thrombin. In patients with

VICC where factor deficiencies prevent clot formation, there still remains active thrombin,

due to activation by the prothrombin activator. Therefore, the substrate in POC machines is

still able to be cleaved, and activated, incorrectly indicating clot formation. This means that

in patients with VICC a POC INR will often give a normal INR result when in fact the

PT/INR is unrecordable when repeated in the laboratory [18, 19]. Table 3 summaries the

value for the above investigations in the diagnosis of VICC.


ACCEPTED MANUSCRIPT

Treatment

The aim of treatment in VICC is to prevent major haemorrhage and complications of this.

This is achieved by neutralising the procoagulant toxins and allowing the recovery of clotting

T
factors to a level where minor trauma does not result in haemorrhage. In patients with active

P
RI
bleeding clotting replacement may be used to speed the recovery of VICC.

SC
Antivenom

NU
Antivenom has been shown to be highly efficacious in binding venom (toxins) and

neutralising toxic effects, both in vitro and in vivo [20]. However, binding and neutralisation
MA
of the toxins will only prevent VICC if given before it develops (very early). For some toxin

types it appears to stop further consumption in patients who have already developed VICC.
ED

Antivenom will not speed the recovery of clotting function beyond this.
PT

The clinical benefit of antivenom in VICC appears to vary for different snakes. Antivenom

has been shown to have little impact on the clinical course of VICC in Australasian elapid
CE

envenoming. In contrast, there is clear evidence that antivenom improves recovery in Echis

species envenoming [13, 21] and other viper envenoming. In an Australian study, the
AC

recovery of PT/INR was compared between patients who had antivenom administered within

6 hours of the bite and patients who received antivenom more than 6 hours after the bite

[21]. There was no significant difference in the improvement of PT/INR between the two

groups, where one would assume if antivenom was having an effect then there would be

faster improvement in the group treated earlier. This compared to a group treated with fresh

frozen plasma (FFP) in addition to antivenom, in which there was a clear benefit with

improvement to an INR <2 within 10 to 12 hours compared to 24 hours without FFP. This

positive result with FFP strengthens the negative finding in the time to antivenom study [21].
ACCEPTED MANUSCRIPT

The natural duration of VICC from Echis envenoming differs from Australasian elapids, with

a prolonged duration of coagulopathy. A recent study from Africa compared the time to

recovery of clotting times and fibrinogen in 60 patients, 47 who received antivenom and 13

T
who did not [13]. The recovery time to a fibrinogen level more than 1 g/L was 7.5 days in the

P
untreated group compared to 40 hours in the antivenom group which was highly statistically

RI
significant (P<0.0001). There was a similar benefit on the PT and the activated partial

SC
thromboplastin time (aPTT). Although this was not a randomised controlled trial the large

treatment effect and objective laboratory outcomes provides strong evidence for the benefit of

NU
antivenom in VICC due to Echis envenoming.
MA
There have been no placebo randomised control studies comparing the effectiveness of

antivenom in VICC and these would be difficult to undertake for ethical reasons. Rather,
ED

there has been numerous randomised controlled trials of antivenom in VICC which have

compared one antivenom to another, different doses or repeat dosing of antivenom and
PT

antivenom to antivenom and heparin or immunoglobulin [16, 22-45]. Further research into
CE

the effectiveness of antivenom for each individual snake group is needed because there are

clear differences in the response of VICC to antivenom.


AC

A major consideration in treating snake envenoming is that VICC rarely occurs as an isolated

envenoming syndrome, but rather, the patient will usually develop either local injury,

myotoxicity or neurotoxicity depending on the snake involved. Because antivenom may be

effective for other clinical effects the reason for antivenom may not always simply be the

presence of VICC. For example, although the administration of antivenom may not change

the clinical course of VICC in Australasian elapid envenoming, early use of antivenom may

prevent neurotoxicity and death due to respiratory paralysis in taipan bites or myotoxicity in
ACCEPTED MANUSCRIPT

tiger snake envenoming [46]. In this case the early presence of abnormal clotting tests are an

excellent indicator for antivenom administration to prevent other effects.

Clotting factor replacement

P T
Fresh Frozen Plasma

RI
Once the procoagulant toxins have been neutralised, clotting function will take 24 to 48 hours

SC
to recover. During this period the patient is at risk of haemorrhage and arguably the most

NU
important intervention is to prevent trauma. For recovery of clotting function to occur more

rapidly there must be exogenous replacement of clotting factors.


MA
A recent randomised controlled trial in Australia of FFP in VICC showed that FFP (10 to

15ml/kg up to maximum of 4 units, about 1000ml) given within 4 hours of antivenom


ED

administration resulted in a more rapid recovery in the PT/INR [47]. The study was too small
PT

to identify a reduction in clinically important outcomes such as major haemorrhage and

death, and the administration of FFP did not decrease the hospital length of stay. Further
CE

larger studies, including studies of different snakes, are required to develop a better

understanding of the potential benefits and risks of FFP. Until further evidence is available
AC

the use of FFP should be confined to patients with active bleeding.

The Australian FFP trial, FFP given within 6 hours of the bite was associated with ongoing

reductions in fibrinogen, despite prior neutralisation with antivenom. The clinical impact of

this remains uncertain, but it does appear that FFP could still "add fuel to the fire" if the

clotting pathway remains activated downstream of the (neutralised or inactive) venom effect

for a period of time after the initial envenoming until a degree of equilibrium is restored [47].

Unfortunately formal studies on the use of other factor replacement products, including

prothrombin complex concentrates (3-factor), cryoprecipitate and fibrinogen concentrates are


ACCEPTED MANUSCRIPT

lacking. The use of cryoprecipitate make senses for snakes producing isolated fibrinogen

deficiencies, but patients can recover adequate clotting function from low levels of

fibrinogen. The advantage of FFP is that it contains all of the major clotting factors, including

T
factors V and VIII which appear to be more important in the recovery of VICC at least in

P
Australian elapids [7].

RI
Heparin

SC
Heparin has been suggested as a treatment for VICC for many years based on the idea that an

NU
anticoagulant may counter the procoagulant toxins. There are surprisingly six studies that

have investigated the effectiveness of heparin in VICC, but they are of low quality with small
MA
numbers, unclear outcomes and positive and negative results [27, 34, 36, 38, 42, 48]. These

studies do not provide sufficient evidence for or against the use of heparin.
ED

Other Haematological Consequences of Snake Bite


PT

Anticoagulant Coagulopathy
CE

A less common syndrome related to snake envenoming is an anticoagulant-type


AC

coagulopathy. The Australian black snake (Pseudechis) genus venom has been shown to

inhibit clotting in vitro and cause a mild to moderate elevation of the aPTT [11]. In a series of

seventeen mulga snake envenomings in Australia ten patients developed an anticoagulant

coagulopathy evidenced by an aPTT median rise of 82s (IQR 55-91s) [3]. Four of these

patients had a small INR rise (<3.0) with the rest having an INR within the normal limits.

Fibrinogen and D-dimer levels were normal. No patients developed any significant clinical

bleeding, supporting the idea that this is not a significant coagulopathy. A similar

anticoagulant coagulopathy was also found in red-bellied black snake (P. porphyriacus) bites.
ACCEPTED MANUSCRIPT

Although this anticoagulant coagulopathy is unlikely to be clinically important, any subtle

elevation in aPTT is an early indicator of envenoming and is therefore a useful marker in

identifying envenoming early. This means that antivenom can be given early based on this. In

T
the case of black snake envenoming which can cause myotoxicity, the early administration of

P
antivenom has been shown to prevent myotoxicity [3, 4].

RI
The occurrence of anticoagulant coagulopathies in other snakes remains unclear, mainly

SC
because the 20 minute WBCT is unlikely to be abnormal except in the most severe cases.

NU
Anticoagulant toxins have been identified in a number of snakes, including cobras (Naja

spp.) and the southern copperhead (Agkistrodon genus). The venom of the southern
MA
copperhead contains a protein C activator [6], but does not appear to have any clinical effects

in humans.
ED

Thrombotic Microangiopathy
PT

A proportion of patients who develop VICC will also go on to develop an envenoming


CE

related thrombotic microangiopathy of which the pathogenesis is not completely understood

[5]. This is characterised by thrombocytopenia, microangiopathic haemolytic anaemia and an


AC

acute kidney injury which continues beyond the resolution of VICC and has been noted in a

number of different snake species, including Australasian elapids and many viper species [5,

42, 49-51]. From the information available in published cases, it is difficult to define where

snakebite induced thrombotic microangiopathy fits on the spectrum of micriangiopathys -

thrombotic thrombocytopenic purpura (TTP) or haemolytic uraemic syndrome (HUS) - and it

is likely to be a distinct syndrome. The combination of thrombocytopenia with mainly renal

involvement is unusual.

In the full blown syndrome which occurs in less than 5% of cases patients develop severe

thrombocytopenia and acute anuric renal failure requiring renal replacement therapy for 2 to
ACCEPTED MANUSCRIPT

6 weeks. [5]. However, the majority of cases are less severe and snake bite induced

thrombotic microangiopathy appears to be a spectrum disorder, with some patients only

having a brief period of thrombocytopenia and a moderate rise in creatinine without oliguria.

T
Treatment is supportive in all cases with renal replacement therapy being the most important

P
RI
intervention in severe cases. Plasmapheresis has been used in a number of more severe cases

but does not appear to change the natural course of the thrombocytopenia, anaemia or renal

SC
injury [49]. The lack of benefit of plasmapheresis is probably due to the underlying cause

NU
being different and not related to ADAMTS-13 function or antibodies like in TTP. An

important consideration is that plasmapheresis will remove the antivenom. It is unclear


MA
whether antivenom alters the course of thrombotic microangiopathy, but is potentially

beneficial for any toxin-related injury resulting in the neutralisation and removal of the toxin
ED

to prevent or improve the syndrome.


PT

Conclusion
CE

Haematological effects are arguably the most important clinical consequences of snakebite

and range from the procoagulant coagulopathy VICC to thrombotic microangiopathy. These
AC

haematological syndromes seen with snake envenoming are unique and an understanding of

the mechanisms is essential to accurate investigation and safe treatment. The most important

complication is major haemorrhage which is often fatal so early intervention and treatment of

VICC is the key to good outcomes.


ACCEPTED MANUSCRIPT

Conflict of Interest

There are no conflicts of interest for any of the authors.

TP
Acknowledgements

RI
SC
Geoff Isbister is funded by an NHMRC Clinical Career Development Award.

NU
MA
ED
PT
CE
AC
ACCEPTED MANUSCRIPT

References

1. Kasturiratne, A., et al., The global burden of snakebite: a literature analysis and

T
modelling based on regional estimates of envenoming and deaths. PLoS Med, 2008.

P
5(11): p. e218.

RI
2. Warrell, D.A., Snake bite. Lancet, 2010. 375(9708): p. 77-88.

SC
3. Johnston, C.I., et al., Mulga snake (Pseudechis australis) envenoming: a spectrum of

myotoxicity, anticoagulant coagulopathy, haemolysis and the role of early antivenom

NU
therapy - Australian Snakebite Project (ASP-19). Clin Toxicol (Phila), 2013. 51(5): p.
MA
417-24.

4. Churchman, A., et al., Clinical effects of red-bellied black snake (Pseudechis


ED

porphyriacus) envenoming and correlation with venom concentrations: Australian

Snakebite Project (ASP-11). Med J Aust, 2010. 193(11-12): p. 696-700.


PT

5. Isbister, G.K., Snakebite doesn't cause disseminated intravascular coagulation:

coagulopathy and thrombotic microangiopathy in snake envenoming. Semin Thromb


CE

Hemost, 2010. 36(4): p. 444-51.


AC

6. Lu, Q., J.M. Clemetson, and K.J. Clemetson, Snake venoms and hemostasis. J Thromb

Haemost, 2005. 3(8): p. 1791-9.

7. Isbister, G.K., et al., Factor deficiencies in venom-induced consumption coagulopathy

resulting from Australian elapid envenomation: Australian Snakebite Project (ASP-

10). J Thromb Haemost, 2010. 8(11): p. 2504-13.

8. McCleary, R.J. and R.M. Kini, Snake bites and hemostasis/thrombosis. Thromb Res,

2013. 132(6): p. 642-6.


ACCEPTED MANUSCRIPT

9. Allen, G.E., et al., Clinical effects and antivenom dosing in brown snake (Pseudonaja

spp.) envenoming--Australian snakebite project (ASP-14). PLoS One, 2012. 7(12): p.

e53188.

T
10. Sano-Martins, I.S., et al., Coagulopathy following lethal and non-lethal envenoming

P
of humans by the South American rattlesnake (Crotalus durissus) in Brazil. QJM,

RI
2001. 94(10): p. 551-9.

SC
11. Isbister, G.K., Procoagulant snake toxins: laboratory studies, diagnosis, and

understanding snakebite coagulopathy. Semin Thromb Hemost, 2009. 35(1): p. 93-

103.
NU
MA
12. Hennerici, M.G., et al., Intravenous ancrod for acute ischaemic stroke in the

European Stroke Treatment with Ancrod Trial: a randomised controlled trial. Lancet,
ED

2006. 368(9550): p. 1871-8.

13. Mion, G., et al., Hemostasis dynamics during coagulopathy resulting from Echis
PT

envenomation. Toxicon, 2013. 76: p. 103-9.

14. O'Leary, M.A. and G.K. Isbister, A turbidimetric assay for the measurement of
CE

clotting times of procoagulant venoms in plasma. J Pharmacol Toxicol Methods,


AC

2010. 61(1): p. 27-31.

15. Isbister, G.K., K. Maduwage, and C.B. Page, Antivenom cross neutralisation in a

suspected Asian pit viper envenoming causing severe coagulopathy. Toxicon, 2014.

90:286-90.

16. Otero, R., et al., Efficacy and safety of two whole IgG polyvalent antivenoms, refined

by caprylic acid fractionation with or without beta-propiolactone, in the treatment of

Bothrops asper bites in Colombia. Trans R Soc Trop Med Hyg, 2006. 100(12): p.

1173-82.
ACCEPTED MANUSCRIPT

17. Isbister, G.K., et al., Diagnostic 20-min whole blood clotting test in Russell's viper

envenoming delays antivenom administration. QJM, 2013. 106(10): p. 925-32.

18. O'Rourke, K.M., et al., Point-of-care derived INR does not reliably detect significant

T
coagulopathy following Australian snakebite. Thromb Res, 2013. 132(5): p. 610-3.

P
19. Cubitt, M., et al., Point-of-care testing in snakebite: an envenomed case with false

RI
negative coagulation studies. Emerg Med Australas, 2013. 25(4): p. 372-3.

SC
20. Isbister, G.K., Antivenom efficacy or effectiveness: the Australian experience.

Toxicology, 2010. 268(3): p. 148-54.

21.
NU
Isbister, G.K., et al., Failure of antivenom to improve recovery in Australian snakebite
MA
coagulopathy. QJM, 2009. 102(8): p. 563-8.

22. Abubakar, I.S., et al., Randomised controlled double-blind non-inferiority trial of two
ED

antivenoms for saw-scaled or carpet viper (Echis ocellatus) envenoming in Nigeria.

PLoS Negl Trop Dis, 2010. 4(7): p. e767.


PT

23. Ariaratnam, C.A., et al., An open, randomized comparative trial of two antivenoms for

the treatment of envenoming by Sri Lankan Russell's viper (Daboia russelii russelii).
CE

Trans R Soc Trop Med Hyg, 2001. 95(1): p. 74-80.


AC

24. Cardoso, J.L., et al., Randomized comparative trial of three antivenoms in the

treatment of envenoming by lance-headed vipers (Bothrops jararaca) in Sao Paulo,

Brazil. Q J Med, 1993. 86(5): p. 315-25.

25. Dart, R.C., et al., A randomized multicenter trial of crotalinae polyvalent immune Fab

(ovine) antivenom for the treatment for crotaline snakebite in the United States. Arch

Intern Med, 2001. 161(16): p. 2030-6.

26. Meyer, W.P., et al., First clinical experiences with a new ovine Fab Echis ocellatus

snake bite antivenom in Nigeria: randomized comparative trial with Institute Pasteur

Serum (Ipser) Africa antivenom. Am J Trop Med Hyg, 1997. 56(3): p. 291-300.
ACCEPTED MANUSCRIPT

27. Myint, L., et al., Heparin therapy in Russell's viper bite victims with impending dic (a

controlled trial). Southeast Asian J Trop Med Public Health, 1989. 20(2): p. 271-7.

28. Karnchanachetanee, C., O. Hanvivatvong, and S. Mahasandana, Monospecific

T
antivenin therapy in Russell's viper bite. J Med Assoc Thai, 1994. 77(6): p. 293-7.

P
29. Otero, R., et al., A randomized double-blind clinical trial of two antivenoms in

RI
patients bitten by Bothrops atrox in Colombia. The Regional Group on Antivenom

SC
Therapy Research (REGATHER). Trans R Soc Trop Med Hyg, 1996. 90(6): p. 696-

700.

30.
NU
Otero-Patino, R., et al., A randomized, blinded, comparative trial of one pepsin-
MA
digested and two whole IgG antivenoms for Bothrops snake bites in Uraba,

Colombia. The Regional Group on Antivenom Therapy Research (REGATHER). Am J


ED

Trop Med Hyg, 1998. 58(2): p. 183-9.

31. Otero, R., et al., A randomized blinded clinical trial of two antivenoms, prepared by
PT

caprylic acid or ammonium sulphate fractionation of IgG, in Bothrops and

Porthidium snake bites in Colombia: correlation between safety and biochemical


CE

characteristics of antivenoms. Toxicon, 1999. 37(6): p. 895-908.


AC

32. Otero-Patino, R., et al., Comparative study of the efficacy and safety of two

polyvalent, caprylic acid fractionated [IgG and F(ab')2] antivenoms, in Bothrops

asper bites in Colombia. Toxicon, 2012. 59(2): p. 344-55.

33. Pardal, P.P., et al., Clinical trial of two antivenoms for the treatment of Bothrops and

Lachesis bites in the north eastern Amazon region of Brazil. Trans R Soc Trop Med

Hyg, 2004. 98(1): p. 28-42.

34. Paul, V., et al., Trial of heparin in viper bites. J Assoc Physicians India, 2003. 51: p.

163-6.
ACCEPTED MANUSCRIPT

35. Paul, V., et al., High-dose anti-snake venom versus low-dose anti-snake venom in the

treatment of poisonous snake bites--a critical study. J Assoc Physicians India, 2004.

52: p. 14-7.

T
36. Paul, V., et al., Trial of low molecular weight heparin in the treatment of viper bites. J

P
Assoc Physicians India, 2007. 55: p. 338-42.

RI
37. Sellahewa, K.H., et al., Intravenous immunoglobulin in the treatment of snake bite

SC
envenoming: a pilot study. Ceylon Med J, 1994. 39(4): p. 173-5.

38. Shah, P.K., et al., Role of heparin in the management of snake (Echis carinatus) bite

NU
cases. J Assoc Physicians India, 1986. 34(9): p. 621-3.
MA
39. Smalligan, R., et al., Crotaline snake bite in the Ecuadorian Amazon: randomised

double blind comparative trial of three South American polyspecific antivenoms.


ED

BMJ, 2004. 329(7475): p. 1129.

40. Thomas, P.P. and J. Jacob, Randomised trial of antivenom in snake envenomation
PT

with prolonged clotting time. Br Med J (Clin Res Ed), 1985. 291(6489): p. 177-8.

41. Warrell, D.A., et al., Bites by the saw-scaled or carpet viper (Echis carinatus): trial of
CE

two specific antivenoms. Br Med J, 1974. 4(5942): p. 437-40.


AC

42. Warrell, D.A., H.M. Pope, and C.R. Prentice, Disseminated intravascular coagulation

caused by the carpet viper (Echis carinatus): trial of heparin. Br J Haematol, 1976.

33(3): p. 335-42.

43. Warrell, D.A., et al., Comparison of Pasteur and Behringwerke antivenoms in

envenoming by the carpet viper (Echis carinatus). Br Med J, 1980. 280(6214): p. 607-

9.

44. Warrell, D.A., et al., Randomized comparative trial of three monospecific antivenoms

for bites by the Malayan pit viper (Calloselasma rhodostoma) in southern Thailand:

clinical and laboratory correlations. Am J Trop Med Hyg, 1986. 35(6): p. 1235-47.
ACCEPTED MANUSCRIPT

45. Jorge, M.T., et al., A randomized 'blinded' comparison of two doses of antivenom in

the treatment of Bothrops envenoming in Sao Paulo, Brazil. Trans R Soc Trop Med

Hyg, 1995. 89(1): p. 111-4.

T
46. Lalloo, D.G., et al., Snake bites by the Papuan taipan (Oxyuranus scutellatus canni):

P
paralysis, hemostatic and electrocardiographic abnormalities, and effects of

RI
antivenom. Am J Trop Med Hyg, 1995. 52(6): p. 525-31.

SC
47. Isbister, G.K., et al., A randomized controlled trial of fresh frozen plasma for treating

venom-induced consumption coagulopathy in cases of Australian snakebite (ASP-18).

NU
J Thromb Haemost, 2013. 11(7): p. 1310-8.
MA
48. Tin Na, S., et al., Heparin therapy in Russell's viper bite victims with disseminated

intravascular coagulation: a controlled trial. Southeast Asian J Trop Med Public


ED

Health, 1992. 23(2): p. 282-7.

49. Isbister, G.K., et al., Thrombotic microangiopathy from Australian brown snake
PT

(Pseudonaja) envenoming. Intern Med J, 2007. 37(8): p. 523-8.

50. Schneemann, M., et al., Life-threatening envenoming by the Saharan horned viper
CE

(Cerastes cerastes) causing micro-angiopathic haemolysis, coagulopathy and acute


AC

renal failure: clinical cases and review. QJM, 2004. 97(11): p. 717-27.

51. Chugh, K.S., et al., Acute renal failure following snakebite. Am J Trop Med Hyg,

1975. 24(4): p. 692-7.

52. Lincz, L.F., et al., Comparative sensitivity of commercially available aPTT reagents

to mulga snake (Pseudechis australis) venom. Pathology, 2014. 46(5): p. 444- 9.


ACCEPTED MANUSCRIPT

Figure Legend:

Figure 1: A schematic representation of where each type of procoagulant toxin acts on the
clotting pathway. Square shapes are inactivate factors and hexagonal shapes are activated
clotting factors. The blue arrows indicate conversion of inactive to active factors and the red
arrows indicate inactivation of the activated clotting factor.

T
Figure 2: A schematic representation of the clotting pathway and the difference between

P
point of care (POC) international normalised ratio (INR) testing and a laboratory INR.

RI
SC
NU
MA
ED
PT
CE
AC
ACCEPTED MANUSCRIPT

TP
RI
SC
NU
MA
ED
PT

Figure 1
CE
AC
ACCEPTED MANUSCRIPT

TP
RI
SC
NU
MA
ED
PT

Figure 2
CE
AC
ACCEPTED MANUSCRIPT

Table 1: A list of snakes grouped by mechanism of coagulopathy, including clotting factor studies in
human cases

Toxin Snake species Common name Factor and haematological


effects
Prothrombin Echis carinatus Saw-scaled viper Fibrinogen, FII, FV, FVIII

T
Activators Echis coloratus Painted carpet viper
Echis ocellatus West African carpet

P
Echis pyramidum viper
Northeast African carpet

RI
viper

SC
Prothrombin Pseudonaja spp. Brown snake Fibrinogen, FII, FV, FVIII
VENOM INDUCED CONSUMPTION COAGULOPATHY

Activators Notechis scutatus Tiger snake


Tropidechis Rough-scaled snake
carinatus Broad-headed snakes

NU
Hoplocephalus Coastal taipan
spp.
Oxyuranus
scutellatus
MA
FX activator Vipera aspis European asp/Asp viper Fibrinogen

FV/FX Daboia russelii Russell’s viper Fibrinogen, FV, FX


ED

activators Daboia russelii Eastern/Siamese


siamensis Russell’s viper
TLEs Atheris spp. Bush vipers Fibrinogen
Bitis arietans African puff adders
PT

Bitis gabonica Gaboon viper


Calloselasma Malayan pit viper
rhodostoma White-lipped green pit
CE

Trimeresurus viper
albolabris Large-eyed pit viper
Trimeresurus Bushmasters
macrops Western diamondback
AC

Lachesis spp. rattlesnake


Crotalus atrox Timber rattlesnake
Crotalus horridus Sahara sand viper
Cerastes vipera

TLEs/Other Cerastes cerastes Saharan horned viper Fibrinogen, FV


Crotalus durissus South American Fibrinogen, FII, FV
Crotalus rattlesnake Fibrinogen, D-dimer
adamanteus Eastern diamondback (normal)
rattlesnake

TLE, PTA, Bothrops jararaca Jararaca Fibrinogen, FII, FV, FVIII


FX Bothrops atrox Common Lancehead Fibrinogen
TLE, FX, FV Bothrops asper Lancehead, Terciopelo Fibrinogen, FII, FV
TLE, PTA
ACCEPTED MANUSCRIPT

Inhibitory Pseudechis Mulga Elevated aPTT


action against australis Red-bellied black snake
ANTICOAGULANT TYPE

FX, FII and Pseudechis Other black snakes


platelets porphyiacus
Pseudechis spp.

P T
Protein C Agkistrodon Southern Copperhead Unknown
activator

RI
SC
NU
MA
ED
PT
CE
AC
ACCEPTED MANUSCRIPT

Table 2: Clinical effects of snakebite induced haemorrhage

Bleeding Syndrome Clinical Effects


Minor bleeding Gingival bleeding
Intravenous cannula site bleeding
Bite site bleeding

T
Haematuria
Epistaxis

P
Gastrointestinal haemorrhage Vomiting frank blood

RI
PR bleeding
Falling Hb
Intracranial haemorrhage Falling GCS

SC
Focal Neurology
Can occur spontaneously, although often in the setting
of hypertension

NU
Traumatic haemorrhage Bleeding from lacerations/wounds
Intracranial bleeding post fall/collapse/syncope
Mediastinal bleeding chest wound/fractured ribs
MA
Long bone cavity bleeding post fractures
Major haemorrhage Falling Hb requiring blood replacement
ED
PT
CE
AC
ACCEPTED MANUSCRIPT

Table 3. The range of laboratory and bedside investigation used for the diagnosis and
monitoring of VICC.

Assay Type Value in assisting diagnosis of VICC

T
WBCT (bedside) Non-standardised test with a low sensitivity [17], but may be of
use if performed under strict guidelines with a 5mL glass tube.

P
Prothrombin time (PT) Most useful diagnostic test in VICC.

RI
or international Unrecordable prothrombin time (INR) indicates complete VICC
normalised ratio (INR) while an INR < 3 occurs with partical VICC. [7] An INR is
- laboratory based useful initially to make the diagnosis and also subsequently to

SC
monitor the recovery of VICC.

INR (Point of care) Not to be used in VICC. False negative results in snake bite due

NU
to assay method used [18].
aPTT Will be abnormal or unrecordable in VICC but adds no further
information to the PT/INR. Useful in anticoagulant
MA
coagulopathy where it will be elevated while the INR is often
normal. However, low phospholipid containing reagents must be
used because high phospholipid content will be insensitive [52].
D-dimer Will be abnormal with any form of VICC but much higher with
ED

prothrombin activators. Should be at least 10 times upper limit


to be considered abnormal in VICC.
May only be mildly elevated in thrombin-like enzyme effects.
Fibrinogen degradation An older test that may be proportionally higher than the D-dimer
PT

products (FDP) with thrombin-like enzymes i.e. 50-100 times normal.


Fibrinogen Undetectable fibrinogen indicates VICC and highly sensitive
clottable assays are the best test to define VICC (i.e. research
CE

setting). However, adds little to an abnormal INR in the clinical


setting except for very mild cases. Fibrinogen is the slowest
recovering factor making it less useful for monitoring recovery
AC

of VICC in the clinical setting.


Other factor assays Specific factor assays can help determine the type of
procoagulant toxins, e.g. low factor X suggests Russell’s viper
envenoming. However, only useful in a research environment or
rarely for exotic snake envenoming.

Vous aimerez peut-être aussi