You are on page 1of 12

British Journal of Anaesthesia 111 (S1): i50–i61 (2013)

doi:10.1093/bja/aet378

Monitoring and managing hepatic disease in anaesthesia


D. Kiamanesh, J. Rumley and V. K. Moitra*

Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
* Corresponding author. E-mail: vm2161@columbia.edu

Summary. Patients with liver disease have multisystem organ


Editor’s key points dysfunction that leads to physiological perturbations ranging
† Hepatic disease can lead to multisystem organ from hyperbilirubinaemia of no clinical consequence to severe
dysfunction with multiple significant anaesthetic coagulopathy and metabolic disarray. Patient-specific risk
implications. factors, clinical scoring systems, and surgical procedures
stratify perioperative risk for these patients. The anaesthetic
† Pathophysiological alterations can occur in
management of patients with hepatic dysfunction involves
pharmacological responses, the systemic and pulmonary
consideration of impaired drug metabolism, hyperdynamic
circulations, coagulation, and neurological function.
circulation, perioperative hypoxaemia, bleeding, thrombosis,
† Perioperative risk is determined by both patient-specific and hepatic encephalopathy.
and procedural factors.
Keywords: liver; liver, cirrhosis; liver, disease; liver, function; liver,
metabolism

The liver metabolizes drugs, food, and toxins; synthesizes pro- (82%), and Class C patients (82%) who undergo abdominal sur-
coagulants and anticoagulants; and regulates temperature, geries.12 13 A newer study reports lower mortality rates after
glucose, and metabolism. When patients with liver disease abdominal operations: Class A¼2%; Class B¼12%; Class
need surgery, anaesthesiologists face several perioperative C¼12%.14 Improvements in surgical technique, anaesthetic
challenges. Patients with liver disease have physiological per- management, and perioperative care might explain the differ-
turbations that range from mild hyperbilirubinemia of no clin- ence in these findings.
ical consequence to severe coagulopathy and metabolic The model for end-stage liver disease (MELD) score is a vali-
disarray (Fig. 1). This article reviews the current body of knowl- dated and prognostic tool that estimates disease severity and
edge of perioperative assessment, monitoring, and manage- survival in patients with liver disease. The MELD score is object-
ment of hepatic disease in patients who will undergo surgery. ive, ranges from 6 to 40 points, and is calculated from the
serum bilirubin, serum creatinine, INR, and aetiology of liver
disease.15 The MELD score has been validated across a
Perioperative risk assessment number of liver diseases (alcoholic hepatitis and variceal
Patients with liver disease who undergo non-hepatic surgery bleeding)16 and surgeries (abdominal, orthopaedic, and car-
have greater transfusion requirements, high infection risk, diovascular).5 8 17 – 19 In patients who have undergone abdom-
cardiac compromise, longer hospital stays, and increased opera- inal surgery, an elevated MELD score was a better predictor of
tive mortality.1 – 6 Patient-specific risk factors, various scoring poor perioperative outcome than CTP classification.17 Patients
systems, and surgical procedures stratify risk for these with MELD scores .15 should avoid elective surgery.20 In
patients.6 – 9 Independent risk factors for complications and 2002, the United Network for Organ Sharing adopted the
mortality are outlined in Table 1. MELD score as a method of allocation of organs to estimate sur-
The Child– Turcotte– Pugh (CTP) classification system, ori- vival. The incorporation of hyponatraemia, an important prog-
ginally devised in 1964 to predict outcomes in cirrhotic patients nostic factor in patients with cirrhosis, into the MELD score
undergoing portosystemic surgery, assesses perioperative risk might improve risk stratification among patients awaiting
for patients with liver disease who undergo hepatic or non- liver transplantation.21
hepatic surgery.8 10 Scores are calculated from five variables: The type of surgical procedure influences perioperative out-
serum albumin, serum bilirubin, ascites, prothrombin time comes in patients with hepatic disease. During abdominal
(PT), or INR (modified CTP score), and grade of encephalopathy surgery, reflexive systemic hypotension from visceral traction
to stratify patients into three groups (Class A¼score 5–6; Class and blood loss can reduce hepatic arterial blood flow.22 23
B¼score 7–9; Class C¼score 10–15).11 This scoring system Patients with extensive liver disease who undergo abdominal
relies on a subjective assessment of ascites and encephalop- surgery have a mortality rate of 30%.12 24 Mortality rates in
athy and does not consider preoperative infection, aetiology cirrhotic patients who undergo open cholecystectomy are
of cirrhosis, or surgery type.6 Original studies suggested high 23–50%; cardiac surgery, 16 –31%; laparotomy for trauma,
mortality rates in Class A patients (10%), Class B patients 45%; and oesophageal surgery, 17– 26%.25 – 33 Patients with

& The Author [2013]. Published by Oxford University Press on behalf of the British Journal of Anaesthesia. All rights reserved.
For Permissions, please email: journals.permissions@oup.com
Monitoring and managing hepatic disease in anaesthesia BJA

Renal/electrolytes/glucose
Acute kidney injury
Hepatorenal syndrome
Hyponatraemia
Hypoglycaemia
Insulin resistance

Hepatic disease
Cardiovascular Decreased metabolic function
Vasodilated state Decreased gluconeogenesis Neurologic
Hyperdynamic cardiac function Decreased lactate clearance Hepatic encephalopathy
Cirrhotic cardiomyopathy Hyperbilirubinemia Cerebral oedema
Drug-induced hypertension Varices Intracranial hypertension
Portopulmonary hypertension Portal hypertension
Ascites

Haematologic
Bleeding and thrombosis
Unreliable INR
Pulmonary Decreased factor synthesis
Aspiration risk Thrombocytopenia and platelet dysfunction
Hepatopulmonary syndrome Dysfibrinogenaemia
Compression atelectasis
Pleural effusions/hepatic hydrothorax
Acute respiratory distress syndrome

Fig 1 Organ response to hepatic dysfunction.

Table 1 Perioperative risk factors associated with complications coagulopathy or CTP Class C cirrhosis are at even higher risk
and mortality1 – 33 during open cholecystectomies, abdominal surgeries, and
cardiovascular procedures.25 27 29 30 32 Contraindications to
Scoring system
elective surgery include fulminant hepatic failure, acute viral
† Modified Child –Turcotte –Pugh (CTP)
† Model for end-stage liver disease (MELD)
or alcoholic hepatitis, chronic active symptomatic hepatitis,
† ASA-PS IV or V CTP Class C cirrhosis, severe coagulopathy (platelet
Patient-specific risk factors count,50 000 ml21, uncorrectable PT .3 s above control),
† Male gender hypoxaemia, heart failure, and acute kidney injury.3 34
† Age .70 yr
† Preoperative infection
† Cirrhosis other than primary biliary cirrhosis Management of anaesthetic medications
† Elevated creatinine and metabolism
† Chronic obstructive pulmonary disease
† Intraoperative hypotension The metabolic activity and clearance of anaesthetic medica-
† Upper gastrointestinal bleeding tions is impaired in patients with liver disease. Without
† Ascites adjusted doses of medications, duration of drug action is
High-risk surgeries extended and toxic levels can result.35 36 Emergence from an-
† Abdominal surgery aesthesia can be delayed.
† Open cholecystectomy
Hepatic metabolism takes place via two pathways: phase 1,
† Cardiothoracic surgery
† Trauma laparotomy biotransformation via the cytochrome P450 system (which
† Oesophageal surgery requires delivery of oxygen via hepatic perfusion); and phase 2,
† Emergency surgery glucuronide conjugation. The first is decreased in liver disease;
the second is relatively spared when dysfunction begins.37

i51
BJA Kiamanesh et al.

Many anaesthetic drugs (i.e. opioids, barbiturates) bind to can develop.58 – 60 Delayed epidural catheter removal has been
albumin in the circulation. Low albumin levels increase the suggested in patients who undergo hepatectomy.61
free fraction of drug and can theoretically exaggerate pharma-
cological effect.38 End-stage liver disease alters hepatic extrac- Organ response, assessment, and
tion, renal excretion, and levels of serum proteins in addition to management
albumin. The net effect of these changes is unpredictable,
making general recommendations for drug administration diffi- Hepatic response
cult. For safety, drugs are titrated to desired effect. Laboratory tests, such as albumin and bilirubin, are non-specific
Patients with hepatic dysfunction can be sensitive to seda- and can be affected by other diseases and medications. Even
tives such as midazolam and diazepam.39 40 In contrast, the with hepatic disease, patients can have normal liver function
pharmacokinetics of lorazepam, which undergoes glucuronida- and laboratory values because the liver, the largest organ and
tion, is less likely to be affected by liver disease.41 Benzodiaze- gland in the body, has a large reserve capacity.
pines can precipitate hepatic encephalopathy, and flumazenil Albumin, produced exclusively in the liver, has been used to
can reverse this condition.42 Conversely, patients who have a assess hepatic synthetic function.62 Low levels of this non-
history of alcohol abuse can require large doses of benzodiaze- specific marker are also found in patients with malnutrition,
pines. Rapid redistribution of bolused propofol determines its renal disease, and capillary leak syndrome. This prognostic
duration of action, which is not affected by impaired liver metab- marker in hospitalized patients is a negative acute phase react-
olism. In a small study of cirrhotic patients, propofol infusions ant because the production of albumin decreases during states
did not increase termination half-life or clearance to a clinically of inflammation with cytokine release and surgical tissue
significant degree.43 trauma.62 63 Albumin, with a long half-life of 20 days, can be
The adverse effects of morphine can be more pronounced normal even in acute liver failure.64
in patients with severe liver disease.44 45 Morphine is metabo- The breakdown of red blood cells within the reticuloendo-
lized by glucuronidation in the liver, and its intermediate thelial system forms unconjugated (‘indirect’) bilirubin. Uncon-
metabolites are eliminated renally. Although the pharmaco- jugated bilirubin is transported to the liver to be conjugated
kinetics of morphine can be unaltered in patients with mild and secreted into bile.64 65 Increased production, decreased
hepatic disease, a prolonged half-life is associated with a pro- hepatic uptake, and decreased conjugation of bilirubin
longed PT, hypoalbuminaemia, encephalopathy, ascites, and cause unconjugated hyperbilirubinaemia. The most common
jaundice.37 46 47 The elimination of morphine metabolites can causes of unconjugated hyperbilirubinaemia are haemolysis,
be prolonged in patients with hepatorenal syndrome (HRS). resolution of haematomas, inborn errors of bilirubin conjuga-
Metabolism of fentanyl, sufentanil, and morphine, which tion (Gilbert’s syndrome and Crigler –Najjar syndrome), or
have a high hepatic extraction ratio, is slowed by impaired portal hypertension.64 Under normal circumstances, levels of
hepatic blood flow.38 44 48 conjugated bilirubin in serum are negligible after rapid excre-
In patients with liver disease and oedema, neuromuscular tion into the biliary system. Conjugated hyperbilirubinaemia
blocking agent doses must be augmented because fluid reten- can indicate intrinsic liver disease, especially in conjunction
tion increases the volume of distribution. Neuromuscular with elevated aminotransferase levels, a sign of hepatocellular
blocking agents that are metabolized by the liver, such as injury. Conjugated hyperbilirubinaemia with elevated alkaline
vecuronium and rocuronium, have a slower onset and longer phosphatase and minimal increase in aminotransferase
duration of action.49 – 54 Liver disease reduces plasma cholin- levels is consistent with a purely cholestatic process and can
esterase activity, and prolonged neuromuscular block after result from numerous causes, including cholestatic drug reac-
succinylcholine has been reported.55 Acute kidney injury with tions or obstructive jaundice.64 – 67
elevated potassium levels or immobilization for long periods The normal portosystemic arterial pressure gradientbetween
might preclude the use of succinylcholine. the portal vein and hepatic vein is ,5 mm Hg. Intrahepatic vas-
Vasodilating anaesthetic agents such as propofol and vola- cular resistance and portal venous blood flow increase the gra-
tile anaesthetics cause hypotension if intravascular volume is dient, characteristic of portal hypertension. Vascular resistance
depleted (i.e. by overdiuresis or gastrointestinal losses), and in the liver can result from several factors: liver fibrosis, re-
reduce liver perfusion. In addition to perioperative haemor- generative nodules, collagen deposits in the space of Disse,
rhage and hypotension, volatile anaesthetic agents can endothelial dysfunction, imbalance between endogenous
reduce hepatic blood flow and hepatic oxygen uptake. Com- vasoconstrictors (i.e. norepinephrine, thromboxane A, endothe-
pared with the vasoconstrictive effects of halothane, isoflur- lin I, angiotensin II, and leukotrienes) and vasodilators (nitric
ane and desflurane can improve hepatic blood flow and oxide), and contraction of portal and septal myofibroblasts
splanchnic perfusion via hepatic vasodilation.56 57 and smooth muscle cells.68 – 70 As the pressure gradient
Patients with end-stage liver disease often exhibit coagula- between the portal vein and the hepatic vein increases, gastro-
tion disorders. The presence of a coagulopathy is a contraindica- oesophageal and haemorrhoidal varices develop to decompress
tion to regional anaesthesia, particularly epidural anaesthesia. the portal system. Portal hypertension produces endogenous
In patients without a preoperative disorder of coagulation, vasodilators that cause splanchnic vasodilation.68 69 71 Splanch-
intraoperative or postoperative coagulopathy from hepatic is- nic vasodilation in turn increases portal blood flow and exacer-
chaemia, insufficient residual hepatic parenchyma, and dilution bates portal hypertension.

i52
Monitoring and managing hepatic disease in anaesthesia BJA
Complications of portal hypertension include gastrointestinal to splanchnic vasodilation and decreased systemic vascular re-
bleeding from expansion and rupture of varices, intraoperative sistance, the body retains sodium and water, which increases
bleeding from intra-abdominal collaterals, and formation of plasma volume. An increase in cardiac output and heart rate
ascites and pleural effusions.68 72 Almost half of patients with cir- follow.81 84 Venous capacitance increases from formation of por-
rhosis have oesophageal varices at the time of diagnosis, with tosystemic shunts with portal hypertension to contribute to a
the 1 yr risk of variceal haemorrhage at 12% and a 1 yr rate of re- hyperdynamic circulation.
current haemorrhage at 60%. The 6 week mortality with each Management of vasodilatory hypotension focuses on
episode of variceal haemorrhage is between 15% and 20%.73 expanding the intravascular volume, administering a vasocon-
In patients with acute upper gastrointestinal bleeding, a restrict- stricting agent such as norepinephrine or vasopressin, and de-
ive transfusion strategy (transfusion when the haemoglobin is termining the underlying cause of vasodilation to target.
,7 g dl21) can decrease subsequent bleeding and prevent an in- Aggressive fluid resuscitation without assessment of fluid re-
crease in the portal-pressure gradient.74 Patients who have sponsiveness should be avoided because excessive i.v. fluid ad-
received multiple previous transfusions, such as patients with a ministration to a non-fluid-responsive patient can increase
history of gastrointestinal bleeding, should be evaluated in cardiac filling pressures, which can cause hepatic congestion,
advance for cross-matching large quantities of blood products pulmonary oedema, and resulting respiratory failure.
possibly required for a procedure. Systemic conditions such as haemochromatosis (ventricular
Before operation, ascites is initially managed with a low hypertrophy with increased end-diastolic and end-systolic
sodium diet and diuretic therapy. Patients with ascites refrac- volumes), amyloidosis (restrictive cardiomyopathy), Wilson’s
tory to treatment with diuretics require large-volume paracen- disease (supraventricular extrasytolic beats), and alcoholism
tesis and albumin replacement to prevent hypotension and (systolic and diastolic dysfunction) can affect liver and cardiac
HRS.75 Large-volume abdominal ascites can lead to a lower function.85 During the perioperative period, the circulatory
venous return and, in conjunction with pleural effusions, can system can be challenged with shunt insertion or greater after-
decrease pulmonary reserve and prevent patients from lying load from surgical stress, unmasking underlying cirrhotic cardio-
flat. If there is evidence of tense ascites, recent food ingestion, myopathy. Echocardiography shows impaired myocardial
or a propensity for delayed gastric emptying, a rapid-sequence function similar to that found in sepsis.86 This condition is also
induction of general anaesthesia with tracheal intubation characterized by QT prolongation and systolic and diastolic dys-
should be considered. function.87 Reduced b-adrenoceptor function might explain
A transjugular intrahepatic portosystemic shunt (TIPS) pre- these findings.85 Cirrhotic cardiomyopathy is often reversible
vents recurrent variceal bleeding and manages refractory and requires inotropic and diuretic support to prevent elevation
ascites.76 During the procedure, an expandable stent is placed of central venous pressure and hepatic congestion.
into the liver parenchyma for portosystemic exchange to de- Patients with portal hypertension can develop portopulmon-
crease portal hypertension. The procedure is performed under ary hypertension (PPHTN) and right ventricular dysfunction.
local or general anaesthesia as a ‘bridge’ to liver transplantation While the exact pathogenesis of PPHTN remains unclear, histo-
in patients with severe liver disease.77 General anaesthesia pathological findings are indistinguishable from those observed
might be preferred because of the duration of the procedure in idiopathic pulmonary artery hypertension (medial hyper-
and the potential for a life-threatening event such as bleeding, trophy, concentric intimal proliferation, plexiform lesions, in
infection, haemobilia, arrhythmias, liver ischaemia, and stent situ thrombosis, and fibrotic changes).88 89 A hyperdynamic cir-
migration.78 Preoperative evaluation of cardiac function (par- culation might increase shear stress on the pulmonary vascula-
ticularly right ventricular function) is essential because stent ture to cause remodelling and pulmonary hypertension.88 90 The
placement can rapidly increase venous return and cardiac mean pulmonary artery pressure .25 mm Hg at rest and pul-
output to unmask underlying cardiomyopathy.79 80 Encephal- monary vascular resistance .240 dyn s cm25 define PPHTN.
opathy can worsen from enhanced systemic delivery of biogenic PPHTN has been reported in 6–9% of patients with advanced
amines normally cleared by the liver. liver disease.91 92 In a retrospective study of patients who
underwent orthotopic liver transplantation (OLT), mortality
was 50% in patients with the mean pulmonary artery pressures
Cardiovascular response between 35 and 50 mm Hg and 100% in patients with pressures
The cardiovascular system of patients with end-stage liver .50 mm Hg.93 Calcium channel blockers, which can be used in
disease mimics the hyperdynamic circulatory changes in patients with pulmonary arterial hypertension, are not recom-
patients with sepsis. Tachycardia, elevated cardiac output, low mended in PPHTN because they produce mesenteric vasodilata-
arterial pressure, and low systemic vascular resistance are char- tion and worsen portal hypertension. b-Blocker therapy, which
acteristic.81 Enhanced endogenous production or diminished is often used in patients with portal hypertension to reduce
hepatic clearance of vasodilating substances (nitric oxide, the risk of variceal bleeding, is contraindicated in patients
carbon monoxide, and endogenous cannabinoids, tumour ne- with PPHTN because its negative inotropic effect decreases
crosis factor-a, adrenomedullin, and hydrogen sulfide) and the exercise tolerance.88 90 Finally, while liver transplantation is a
inflammatory response to bacterial translocation cause tempting option to reduce PPHTN, moderate or severe uncon-
splanchnic arterial vasodilation (Fig. 2).81 82 These changes trolled PPHTN is associated with significant mortality risk and
have been reversed with liver transplantation.81 83 In response is a contraindication to OLT.93 – 95 Intraoperative management

i53
BJA Kiamanesh et al.

Increased levels of vasodilating substances


Nitric oxide
Carbon monoxide
Prostacyclin
Endocannabinoids
Endothelium-derived hyperpolarizing factor
Tumour necrosis factor-a
Adrenomedullin
Hydrogen sulfide

Liver dysfunction Splanchnic vasodilation


Portal hypertension Systemic vasodilation
Bacterial translocation and inflammation

Increased plasma volume


Increased cardiac output

Fig 2 Vasodilation from hepatic dysfunction.

is targeted to prevent increases in pulmonary vascular resist- chronic pulmonary disease and occasionally acute respiratory
ance by controlling hypoxia, hypercarbia, acidosis, pain, and distress syndrome.99 Chest radiography rules out pulmonary
hypothermia. oedema, pleural effusions, or cardiomegaly in patients with
Because of these cardiovascular changes, invasive monitors symptoms of dyspnoea. Muscle wasting and intra-abdominal
usually are used during the perioperative period. Beat-to-beat hypertension from ascites increase the work of breathing.
arterial pressure is monitored and blood is sampled frequently Ascites fluid can enter the pleural space through small chan-
via a radial artery catheter. Pulse pressure variation, calculated nels in the diaphragm to cause a hepatic hydrothorax (usually
from the invasive arterial tracing, assesses fluid responsive- on the right side). Compressive atelectasis from ascites and
ness. Peripheral arterial pressure measurements can be unre- pleural effusions reduce functional residual capacity and de-
liable in patients with severe vasoconstriction or vasodilation. crease the time to oxygen desaturation. Lung recruitment
Central venous access is established to measure right atrial and pleural fluid drainage can improve pulmonary mechanics.
pressure and to administer drugs. Monitoring central venous Respiratory alkalosis can be observed in patients with a high
pressure does not reliably measure blood volume or change respiratory drive from liver dysfunction. The use of positive-
in blood volume.96 Echocardiography assesses ventricular pressure mechanical ventilation and PEEP can lead to an
filling, contractility, and function. Myocardial ischaemia, pul- increased average intrathoracic pressure, which decreases
monary embolism, pleural effusion, and inadequate inferior venous return from the inferior vena cava and hepatic veins
vena cava reconstruction can be observed with transoesopha- and can lead to congestion of the liver. This concern remains
geal echocardiography (TOE). The risk of rupture of oesopha- controversial, as studies with PEEP of up to 10 cm H2O have
geal varices with TOE is rare.97 98 Bladder pressures should shown no adverse effects on hepatic arterial, portal venous, or
be measured if intra-abdominal hypertension from ascites is hepatic venous flow.101
suspected. A diagnosis of HPS is considered in patients without cardiopul-
monary disease who have a PaO2 ,8.0 kPa. The triad of liver
disease and/or portal hypertension, widened age-corrected al-
Pulmonary response veolar–arterial oxygen gradient (.2–2.7 kPa) while breathing
Up to 70% of patients with end-stage liver disease complain of room air, and documented intrapulmonary vascular dilation
dyspnoea.99 100 Potential causes of preoperative hypoxaemia (IPVD) define HPS.99 102 103 Enhanced production or impaired
and respiratory failure in patients with cirrhosis include atel- hepatic clearance of endogenous vasodilators (i.e. nitric oxide,
ectasis from the compressive effects of ascites, hepatic vasodilator prostaglandins, substance P), or inhibition of vascon-
hydrothorax, hepatopulmonary syndrome (HPS), underlying strictive substances (i.e. tyrosine, serotonin, and endothelin) by a

i54
Monitoring and managing hepatic disease in anaesthesia BJA
damaged livercan cause IPVD. IPVD causes hypoxaemiaviaven- renal function is impaired rapidly and progressively with a doub-
tilation and perfusion mismatching, intrapulmonary shunt ling of the initial serum creatinine to .2.5 mg dl21 or a 50% re-
physiology, and diffusion limitation.99 103 Patients with HPS duction in the 24 h creatinine clearance to ,20 ml min21 in ,2
often complain of dyspnoea and fatigue; they can have clubbing weeks. Patients with type II HRS have impaired renal function
and spider angiomata. They may experience platypnoea (dys- and a serum creatinine .1.5 mg dl21 and do not meet the cri-
pnoea in the upright position relieved by recumbency) or have teria for type I HRS.107 Compared with patients diagnosed with
orthodeoxia (arterial oxyhaemoglobin desaturation in the prerenal failure, patients with HRS lack a renal response to a
upright position). Preferential perfusion of IPVDs with the 1.5 litre volume challenge. Distinguishing between HRS and
patient upright might cause these clinical manifestations. acute tubular necrosis is difficult. Fractional excretion of
Contrast-enhanced echocardiography or perfusion lung scan- sodium ,1% suggests tubular function and favours a diagnosis
ning with technetium-99m-labelled macroaggregate albumin of HRS. The presence of renal tubular epithelial cells in urinary
detects intrapulmonary shunting suggestive of IPVD.102 103 sediment favours a diagnosis of acute tubular necrosis.109 112
Resolution of this syndrome after transplant has been reported. Administration of terlipressin, a vasopressin analogue, along
Medical therapy for HPS has, to date, been relatively ineffective. with i.v. albumin has been shown to have some benefit in
Currently, the only known treatment option for patients with HPS patients with type 1 HRS.114 115 Medical management of HRS
is liver transplantation.104 – 106 is marginally effective, but liver transplantation usually reverses
HRS.116 117
Hypervolaemic hyponatraemia from accumulated secretion
Renal, electrolyte, and glucose response of anti-diuretic hormone acts on vasopressin-2 receptors of
Acute kidney injury commonly occurs in patients with chronic the renal tubular collecting duct to impair excretion of solute-
liver disease and is present in up to 20% of patients hospitalized free water.118 When extracellular volume expands, ascites and
with decompensated liver cirrhosis.107 Patients with acute oedema can result. In patients with hypovolaemic hyponatrae-
kidney injury and cirrhosis have more complications and mia via loss of extracellular fluid from the kidneys (overdiuresis)
greater risk for mortality after liver transplantation than those or gastrointestinal tract, ascites or oedema is rare. These
without renal failure. Gastrointestinal bleeding, diarrhoea from patients can have prerenal failure from low circulating volume
infection or lactulose administration, and diuretic medications or hepatic encephalopathy from rapid reduction in serum
change circulatory function via hypovolaemia and can cause pre- osmolality.109 118 Distinguishing between hypervolaemic and
renal injury. This injury rapidly reverses with correction of the hypovolaemic hyponatremia guides treatment. Hypervolaemic
underlying pathophysiology and volume resuscitation.108 109 hyponatraemia is managed with fluid restriction (1–1.5 litre
As cirrhosis progresses, reduction in systemic vascular resis- day21) and withholding of diuretics. Vaptans, medications
tance activates the renin–angiotensin and sympathetic nervous that block the V2 receptor, increase solute-free water excretion
systems, which leads to ascites, oedema, and vasoconstric- dose-dependently and might preclude water restriction so that
tion of the intrarenal circulation and renal hypoperfusion. diuretics can be continued.119 Patients with hypovolaemic hypo-
Ischaemic events and medications such as non-steroidal anti- natraemia are not given diuretics; instead, saline is administered
inflammatory drugs or aminoglycosides cause intrinsic renal to increase plasma volume.118
damage.108 109 Managing kidney injury, low urine output, and Patients with preoperative hyponatraemia are at risk for
electrolyte abnormalities in patients with hepatic disease is central pontine myelinolysis (CPM) perioperatively. CPM is a
challenging. Pre- or post-renal kidney injuries reverse with neurological condition characterized by symmetric non-
enhanced renal perfusion or removal of the obstruction, re- inflammatory demyelinating lesions in the basis pontis. The aeti-
spectively. Optimizing renal perfusion and avoidance of ology of CPM is uncertain, but osmotic stress on central nervous
nephrotoxic drugs are also important in these patients.110 Con- system cells has been suggested, and CPM correlates with rapid
tinuous renal replacement therapy can be used perioperatively correction of hyponatraemia.118 Rapid changes in concentration
to manage volume shifts, acid– base balance, and electrolyte of serum sodium cannot be predicted and a ‘safe’ rate of cor-
disturbances. Placement of a TIPS can improve serum creatin- rection of hyponatraemia has not been definitively established;
ine, with more profound effects on those patients with higher some experts recommend sodium correction at a rate ,12 mEq
baseline levels.111 litre21 day21.120 – 122 Preoperative correction of hyponatraemia
HRS is caused by functional renal vasoconstriction (via activa- can prevent a rapid increase in serum sodium levels intraopera-
tion of the renin–angiotensin system, the sympathetic nervous tively and after operation. Sodium bicarbonate should be admi-
system, and arginine vasopressin) in response to splanchnic ar- nistered cautiously because it has a high concentration of
terial vasodilation.109 112 Although histological findings and sodium.
diagnostic tests for HRS are lacking, diagnostic criteria are Calcineurin inhibitors, loop diuretics, and significant blood
used to categorize two types of HRS. Criteria for HRS include (i) loss can worsen hypomagnesaemia. Total serum calcium is
cirrhosis with ascites, (ii) serum creatinine .1.5 mg dl21, (iii) often low because patients commonly have low serum
no improvement of creatinine after 2 days of diuretic withdrawal albumin with reduced calcium binding. Failure to clear citrate
and volume expansion with albumin, (iv) absence of shock, (v) (metabolized by the liver) after the administration of a signifi-
no current or recent treatment with nephrotoxic drugs, and cant volume of packed red blood cells, malnutrition, and
(vi) absence of parenchymal kidney disease.113 In type I HRS, vitamin D deficiency can lead to perioperative hypocalcaemia.

i55
BJA Kiamanesh et al.

Disorders of glucose metabolism are observed in patients thromboelastography or rotational thromboelastometry


with chronic liver disease and cirrhosis.123 124 Multiple factors assess the viscoelastic properties of a whole blood sample as a
affect glucose levels, including the inflammatory response to function of time to reflect the function and interaction of
surgery, steroid administration, hepatic dysfunction, altered plasma, blood cells, and platelets. These measurements
glycogen stores, and insulin resistance in liver failure. Episodes detect a hypercoagulable state and distinguish hyperfibrinolysis
of hypoglycaemia are observed in patients with end-stage liver from other causes of coagulopathy, such as factor depletion or
disease. thrombocytopenia to offer a composite picture of the clotting
cascade.129 130 140 141 Although both have been used to guide
haemostatic management during liver transplantation, there
Haematological response are no studies evaluating these tests as predictors of intraopera-
In patients with hepatic disease, haemostatic changes tive bleeding in patients with hepatic disease.129 130 140
promote both bleeding and thrombosis. Inadequate synthesis Uncontrolled haemorrhage and massive transfusion in the
of all coagulation factors (except for von Willebrand’s factor operating theatre or the intensive care unit can cause the
and factor VIII), thrombocytopenia, platelet function defects, lethal triad of acidosis, coagulopathy, and hypothermia. Left
dysfibrinogenaemia, and elevated tissue plasminogen activa- uncorrected, each of these abnormalities can exacerbate the
tor (tPA) levels cause bleeding. Elevation of von Willebrand’s others, creating a ‘bloody vicious cycle’. Additional early com-
factor and Factor VIII and decreased levels of ADAMTS-13 (a plications of massive transfusion include (i) acute haemolytic
disintegrin and metalloproteinase with a thrombospondin transfusion reactions, (ii) febrile non-haemolytic transfusion
type 1 motif, member 13), protein C, protein S, antithrombin, reactions, (iii) transfusion-related acute lung injury (TRALI),
a 2-macroglobulin, plasminogen, and heparin cofactor II (iv) transfusion-associated circulatory overload, (v) allergic
favour thrombosis. reactions, (vi) bacterial sepsis, (vii) hypocalcaemia, and (viii)
Factors VII, X, V, II (prothrombin), and I (fibrinogen) have a hyperkalaemia (see Pham and Shaz,142 this issue).143
short half-life (hours to days) and are synthesized solely by hepa- Warming the room, applying forced warming systems,
tocytes, making possible a ‘semi real-time’ evaluation of hepatic administering blood products and fluids through a fluid
synthetic function.125 Although patients with liver disease can warmer, and heating and humidifying inspired gases reduce
have abnormal PT from decreased production of pro-coagulant the risk of hypothermia. Dilutional coagulopathy and thrombo-
factors, the rebalanced state of pro- and anti-coagulants in cytopenia may be prevented by transfusing packed red blood
patients with liver disease makes the PT an unreliable tool for cells, fresh-frozen plasma, and platelets in a 1:1:1 ratio.143 Occa-
evaluating tendency to bleed or clot.126 – 133 sionally, recombinant Factor VIIa is administered.144 – 148 Factor
Levels of fibrinogen, an acute phase reactant, are normal or VIIa has not been shown to decrease red blood cell transfusion
increased in mild-to-moderate liver disease. In patients with requirements during hepatectomies.149 Potassium, magne-
severe hepatic dysfunction, however, fibrinogen is poorly sium, and calcium levels are frequently monitored to correct ab-
synthesized and dysfunctional, which increases the risk of normal levels. Improving haemodynamic parameters, adjusting
bleeding.134 135 Although hyperfibrinolysis (from increased minute ventilation, or administering sodium bicarbonate or tro-
levels of tPA and reduced levels of thrombin-activatable fibrin- methamine manage metabolic acidosis. The riskof TRALI can be
olysis inhibitor and plasmin inhibitor) has been observed in cir- reduced by minimizing transfusions and transfusing packed red
rhotic patients, its association with bleeding is unclear.126 136 blood cells with a short storage time (see Cohen and Matot,150
Hypofibrinolysis (from increased levels of plasminogen activa- this issue) and fresh-frozen plasma from men or nulliparous
tor inhibitor and reduced levels of plasminogen) can restore the women.
balance of fibrinolysis.126 137 138
Thrombocytopenia and platelet dysfunction are character-
istic of end-stage liver disease. Thrombocytopenia results Neurological response
from several factors: portal hypertension with hypersplenism, Patients with liver disease can have encephalopathy before op-
which sequesters platelets; consumption of platelets during eration or after operation. Brain dysfunction manifests in acute
systemic intravascular coagulation; and impaired hepatic syn- or chronic liver failure or after a spontaneous or surgical porto-
thesis of thrombopoietin, which stimulates platelet production systemic shunt.151 152 Hepatic encephalopathy is a neuro-
in the bone marrow.72 139 Defective signal transduction; psychiatric complication of acute and chronic liver disease
uraemia from acute kidney injury; and intrinsic defects of with features that range from mild confusion to cerebral
ADP, arachidonic acid, collagen, and thrombin prevent platelet oedema with intracranial hypertension.101 152 Patients have
aggregation.139 disturbances in consciousness, cognitive abilities, behaviour,
Patients with active sites of bleeding should be transfused fine motor skills, concentration, reaction time, memory,
with fresh-frozen plasma, packed red blood cells, cryoprecipi- and/or electroencephalogram.152 – 154 The pathogenesis of
tate, or platelets as dictated by clinical assessment and labora- hepatic encephalopathy is not understood, but most theories
tory tests. In addition, desmopressin can be considered for implicate elevated levels of ammonia, a gut-derived neuro-
patients with concomitant renal dysfunction and uraemic toxin, which is shunted to the systemic circulation from the
bleeding. Coagulation status should always be monitored by portal system. Bacteria in the intestinal tract produce
inspection of the surgical field. Measurements from ammonia, which crosses the blood– brain barrier into

i56
Monitoring and managing hepatic disease in anaesthesia BJA
astrocytes that detoxify it to glutamine. An increased concen- Conclusions
tration of intracellular glutamine leads to swollen astrocytes
Patients with hepatic disease are at high risk for poor periopera-
with reduced ability to regulate neurotransmission.80 101 152
155 tive outcomes. Assessment of the severity of liver dysfunction
High serum ammonia levels are present in patients with
and consideration of type of surgery stratifies risk and delays
hepatic encephalopathy, even though the degree of elevation
elective surgery in patients with significant hepatic disease
of ammonia does not correlate with neurological dysfunction.
(CTP Class C cirrhosis). Managing surgical patients with hepatic
Other factors, such as hyponatraemia, gastrointestinal bleed-
disease considers anaesthetic dose adjustment and monitoring
ing, and infection, may contribute to the development of
the multisystem organ responses to liver dysfunction.
hepatic encephalopathy.101 151 156 Left untreated, cerebral
oedema can progress to intracranial hypertension and brain
herniation. Multiple grading scales assess the severity of Authors’ contributions
hepatic encephalopathy. The most common scales are the
All authors participated in literature search, writing of first
West Haven Criteria (Conn Score) which has grades 0 through
draft, editing, and final approval.
4 and the Glasgow coma score.157 158
Initial therapy for hepatic encephalopathy identifies
and treats reversible triggers (i.e. gastrointestinal bleeding, Acknowledgement
hypokalaemia, alkalaemia, hypoglycaemia, hypovolaemia, The authors would like to thank Sally Kozlik for her invaluable
and infection) of this neuropsychiatric syndrome and considers editorial assistance.
minimizing benzodiazepine medications.14 82 While there is no
evidence of a direct correlation between the degree of elevation
of ammonia and severity of hepatic encephalopathy, it is well Declaration of interest
accepted that ammonia is a factor.159 Non-absorbable disac- None declared.
charides such as lactulose acidify the gut environment to de-
crease absorption of ammonia from the gut via catharsis.152
Excessive dosing of lactulose causes dehydration however. References
Oral antibiotics (rifaximin, neomycin, vancomycin, paramomy- 1 Csikesz NG, Nguyen LN, Tseng JF, Shah SA. Nationwide volume and
cin, or metronidazole) reduce ammonia-producing enteric bac- mortality after elective surgery in cirrhotic patients. J Am Coll Surg
teria.156 Rifaximin, in combination with lactulose, might reduce 2009; 208: 96– 103
episodes of hepatic encephalopathy.151 156 2 del Olmo JA, Flor-Lorente B, Flor-Civera B, et al. Risk factors for non-
Patients who develop fulminant hepatic failure are at risk for hepatic surgery in patients with cirrhosis. World J Surg 2003; 27:
647–52
hepatic encephalopathy, cerebral oedema with increased
3 Friedman LS. The risk of surgery in patients with liver disease.
intracranial pressure (ICP), and herniation. In cases of altered
Hepatology 1999; 29: 1617–23
mental status, a head CT scan is often indicated to evaluate
4 Rice HE, O’Keefe GE, Helton WS, Johansen K. Morbid prognostic
intracranial bleeding, herniation, the extent of cerebral features in patients with chronic liver failure undergoing nonhepa-
oedema, or both. ICP monitoring should be seriously consid- tic surgery. Arch Surg 1997; 132: 880–4; discussion 4– 5
ered for patients with fulminant hepatic failure and encephal- 5 Teh SH, Nagorney DM, Stevens SR, et al. Risk factors for mortality
opathy, even given the bleeding risks associated with invasive after surgery in patients with cirrhosis. Gastroenterology 2007;
monitors. Bleeding risk seems to be the greatest deterrent to 132: 1261– 9
ICP monitoring. 6 Ziser A, Plevak DJ, Wiesner RH, Rakela J, Offord KP, Brown DL. Mor-
ICP elevations to .25 mm Hg should be treated with manni- bidity and mortality in cirrhotic patients undergoing anesthesia
and surgery. Anesthesiology 1999; 90: 42–53
tol to achieve hyperosmolarity. Preoperative hyperventilation
7 Cho HC, Jung HY, Sinn DH, et al. Mortality after surgery in patients
has not been shown to improve outcome and corticosteroids
with liver cirrhosis: comparison of Child –Turcotte– Pugh, MELD
are not indicated. Pentobarbital coma can be used for patients
and MELDNa score. Eur J Gastroenterol Hepatol 2011; 23: 51– 9
unresponsive to mannitol, but can worsen cerebral hypo-
8 Farnsworth N, Fagan SP, Berger DH, Awad SS. Child– Turcotte–Pugh
perfusion by causing systemic hypotension. Many centres con- versus MELD score as a predictor of outcome after elective and
sider sustained cerebral hypoperfusion (cerebral perfusion emergent surgery in cirrhotic patients. Am J Surg 2004; 188:
pressure ,40 mm Hg) a contraindication to transplant because 580–3
of the high risk for brain death. In the future, transcranial 9 Hoteit MA, Ghazale AH, Bain AJ, et al. Model for end-stage liver
Doppler ultrasonography, near-infrared spectrophotometry, disease score versus child score in predicting the outcome of sur-
and measurements of extracellular lactate via cerebral micro- gical procedures in patients with cirrhosis. World J Gastroenterol
2008; 14: 1774–80
dialysis could offer means of monitoring for elevated ICP in
10 Cholongitas E, Marelli L, Shusang V, et al. A systematic review of
these patients.160 161 For anaesthesia, drugs or conditions that
the performance of the model for end-stage liver disease (MELD)
exacerbate elevations in ICP should be avoided. Recent evidence
in the setting of liver transplantation. Liver Transpl 2006; 12:
shows that moderate hypothermia (32–338C) can reduce cere- 1049–61
bral oedema and intracerebral hypertension; however, the side- 11 Pugh RNH, Murray-Lyon IM, Dawson JL, Pietroni MC, Williams R.
effects of these manoeuvres are greater risk of infection, cardiac Transection of the oesophagus for bleeding oesophageal
dysrhythmias, and coagulopathy.162 – 166 varices. Br J Surg 1973; 60: 646–8

i57
BJA Kiamanesh et al.

12 Garrison RN, Cryer HM, Howard DA, Polk HC Jr. Clarification of risk 33 Tachibana M, Kotoh T, Kinugasa S, et al. Esophageal cancer with
factors for abdominal operations in patients with hepatic cirrhosis. cirrhosis of the liver: results of esophagectomy in 18 consecutive
Ann Surg 1984; 199: 648 –55 patients. Ann Surg Oncol 2000; 7: 758– 63
13 Mansour A, Watson W, Shayani V, Pickleman J. Abdominal opera- 34 RN S. Anesthetic concerns for the patient with liver disease. Anesth
tions in patients with cirrhosis: still a major surgical challenge. Analg 2008; 106: 1 –7
Surgery 1997; 122: 730– 5; discussion 5–6 35 Prescott LF, Forrest JAH, Adjepon-Yamoah KK, Finlayson NDC. Drug
14 Telem DA, Schiano T, Goldstone R, et al. Factors that predict metabolism in liver disease. J Clin Pathol 1975; 28: 62 –5
outcome of abdominal operations in patients with advanced cir- 36 Howden CW, Birnie GG, Brodie MJ. Drug metabolism in liver
rhosis. Clin Gastroenterol Hepatol 2010; 8: 451– 7, quiz e58 disease. Pharmacol Ther 1989; 40: 439–74
15 Kamath PS, Wiesner RH, Malinchoc M, et al. A model to predict sur- 37 Hoyumpa AM, Schenker S. Is glucuronidation truly preserved in
vival in patients with end-stage liver disease. Hepatology 2001; 33: patients with liver disease? Hepatology 1991; 13: 786– 95
464– 70 38 Delco F, Tchambaz L, Schlienger R, Drewe J, Krahenbuhl S. Dose ad-
16 Said A, Williams J, Holden J, et al. Model for end stage liver disease justment in patients with liver disease. Drug Saf 2005; 28: 529– 45
score predicts mortality across a broad spectrum of liver disease. 39 MacGilchrist AJ, Birnie GG, Cook A, et al. Pharmacokinetics and
J Hepatol 2004; 40: 897 –903 pharmacodynamics of intravenous midazolam in patients with
17 Befeler AS, Palmer DE, Hoffman M, Longo W, Solomon H, Di severe alcoholic cirrhosis. Gut 1986; 27: 190–5
Bisceglie AM. The safety of intra-abdominal surgery in patients 40 Ochs HR, Greenblatt DJ, Eckardt B, Harmatz JS, Shader RI.
with cirrhosis: model for end-stage liver disease score is superior Repeated diazepam dosing in cirrhotic patients: cumulation and
to Child– Turcotte– Pugh classification in predicting outcome. sedation. Clin Pharmacol Ther 1983; 33: 471–6
Arch Surg 2005; 140: 650 –4; discussion 5
41 Kraus JW, Desmond PV, Marshall JP, Johnson RF, Schenker S,
18 Northup PG, Wanamaker RC, Lee VD, Adams RB, Berg CL. Model for Wilkinson GR. Effects of aging and liver disease on disposition of
end-stage liver disease (MELD) predicts nontransplant surgical lorazepam. Clin Pharmacol Ther 1978; 24: 411– 9
mortality in patients with cirrhosis. Ann Surg 2005; 242: 244– 51
42 Bansky G, Meier PJ, Riederer E, Walser H, Ziegler WH, Schmid M.
19 Perkins L, Jeffries M, Patel T. Utility of preoperative scores for pre- Effects of the benzodiazepine receptor antagonist flumazenil in
dicting morbidity after cholecystectomy in patients with cirrhosis. hepatic encephalopathy in humans. Gastroenterology 1989; 97:
Clin Gastroenterol Hepatol 2004; 2: 1123–8 744– 50
20 Hanje AJ, Patel T. Preoperative evaluation of patients with liver 43 Servin F, Cockshott ID, Farinotti R, Haberer JP, Winckler C,
disease. Nat Clin Pract Gastroenterol Hepatol 2007; 4: 266–76 Desmonts JM. Pharmacokinetics of propofol infusions in patients
21 Kim WR, Biggins SW, Kremers WK, et al. Hyponatremia and mortal- with cirrhosis. Br J Anaesth 1990; 65: 177– 83
ity among patients on the liver-transplant waiting list. N Engl J Med 44 Tegeder I, Lotsch J, Geisslinger G. Pharmacokinetics of opioids in
2008; 359: 1018–26 liver disease. Clin Pharmacokinet 1999; 37: 17–40
22 Gelman SI. Disturbances in hepatic blood flow during anesthesia 45 Laidlaw J, Read AE, Sherlock S. Morphine tolerance in hepatic cir-
and surgery. Arch Surg 1976; 111: 881 –3 rhosis. Gastroenterology 1961; 40: 389– 96
23 Torrance HB. Liver blood flow during operations on the upper 46 Hasselstrom J, Eriksson S, Persson A, Rane A, Svensson JO,
abdomen. J Roy Coll Surg Edinb 1957; 2: 216– 28 Sawe J. The metabolism and bioavailability of morphine in
24 Neeff H, Mariaskin D, Spangenberg HC, Hopt UT, Makowiec F. patients with severe liver cirrhosis. Br J Clin Pharmacol 1990; 29:
Perioperative mortality after non-hepatic general surgery in 289– 97
patients with liver cirrhosis: an analysis of 138 operations in the 47 Mazoit JX, Sandouk P, Zetlaoui P, Scherrmann JM. Pharmacokinet-
2000 s using Child and MELD scores. J Gastrointest Surg 2011; ics of unchanged morphine in normal and cirrhotic subjects.
15: 1– 11 Anesth Analg 1987; 66: 293–8
25 Aranha GV, Sontag SJ, Greenlee HB. Cholecystectomy in cirrhotic 48 Devlin JW, Roberts RJ. Pharmacology of commonly used analge-
patients: a formidable operation. Am J Surg 1982; 143: 55– 60 sics and sedatives in the ICU: benzodiazepines, propofol, and
26 Belghiti J, Cherqui D, Langonnet F, Fekete F. Esophagogastrectomy opioids. Crit Care Clin 2009; 25: 431– 49, vii
for carcinoma in cirrhotic patients. Hepato-Gastroenterology 49 Hunter JM, Parker CJ, Bell CF, Jones RS, Utting JE. The use of differ-
1990; 37: 388– 91 ent doses of vecuronium in patients with liver dysfunction. Br J
27 Bloch RS, Allaben RD, Walt AJ. Cholecystectomy in patients with Anaesth 1985; 57: 758– 64
cirrhosis. A surgical challenge. Arch Surg 1985; 120: 669– 72 50 Khalil M, D’Honneur G, Duvaldestin P, Slavov V, De Hys C, Gomeni R.
28 Demetriades D, Constantinou C, Salim A, Velmahos G, Rhee P, Pharmacokinetics and pharmacodynamics of rocuronium in
Chan L. Liver cirrhosis in patients undergoing laparotomy for patients with cirrhosis. Anesthesiology 1994; 80: 1241– 7
trauma: effect on outcomes. J Am Coll Surg 2004; 199: 538– 42 51 Lebrault C, Berger JL, D’Hollander AA, Gomeni R, Henzel D,
29 Douard R, Lentschener C, Ozier Y, Dousset B. Operative risks of di- Duvaldestin P. Pharmacokinetics and pharmacodynamics of
gestive surgery in cirrhotic patients. Gastroenterol Clin Biol 2009; vecuronium (ORG NC 45) in patients with cirrhosis. Anesthesiology
33: 555–64 1985; 62: 601– 5
30 Filsoufi F, Salzberg SP, Rahmanian PB, et al. Early and late outcome 52 Magorian T, Wood P, Caldwell J, et al. The pharmacokinetics and
of cardiac surgery in patients with liver cirrhosis. Liver Transpl neuromuscular effects of rocuronium bromide in patients with
2007; 13: 990– 5 liver disease. Anesth Analg 1995; 80: 754–9
31 Klemperer JD, Ko W, Krieger KH, et al. Cardiac operations in 53 Servin FS, Lavaut E, Kleef U, Desmonts JM. Repeated doses of
patients with cirrhosis. Ann Thorac Surg 1998; 65: 85 –7 rocuronium bromide administered to cirrhotic and control
32 Suman A, Barnes DS, Zein NN, Levinthal GN, Connor JT, Carey WD. patients receiving isoflurane. A clinical and pharmacokinetic
Predicting outcome after cardiac surgery in patients with cirrhosis: study. Anesthesiology 1996; 84: 1092–100
a comparison of Child– Pugh and MELD scores. Clin Gastroenterol 54 van Miert MM, Eastwood NB, Boyd AH, Parker CJ, Hunter JM. The
Hepatol 2004; 2: 719– 23 pharmacokinetics and pharmacodynamics of rocuronium in

i58
Monitoring and managing hepatic disease in anaesthesia BJA
patients with hepatic cirrhosis. Br J Clin Pharmacol 1997; 44: shunt (TIPS) in the management of portal hypertension: update
139– 44 2009. Hepatology 2010; 51: 306
55 Viby-Mogensen J, Hanel HK. Prolonged apnoea after suxameth- 77 DeGasperi A, Corti A, Corso R, et al. Transjugular intrahepatic por-
onium: an analysis of the first 225 cases reported to the Danish tosystemic shunt (TIPS): the anesthesiological point of view
Cholinesterase Research Unit. Acta Anaesthesiol Scand 1978; 22: after 150 procedures managed under total intravenous anesthe-
371– 80 sia. J Clin Monit Comput 2009; 23: 341– 6
56 Gatecel C, Losser MR, Payen D. The postoperative effects of halo- 78 Gaba RC, Khiatani VL, Knuttinen MG, et al. Comprehensive review
thane versus isoflurane on hepatic artery and portal vein blood of TIPS technical complications and how to avoid them. Am J
flow in humans. Anesth Analg 2003; 96: 740 –5, table of contents Roentgenol 2011; 196: 675–85
57 O’Riordan J, O’Beirne HA, Young Y, Bellamy MC. Effects of desflur- 79 Colombato LA, Spahr L, Martinet JP, et al. Haemodynamic adapta-
ane and isoflurane on splanchnic microcirculation during major tion two months after transjugular intrahepatic portosystemic
surgery. Br J Anaesth 1997; 78: 95 –6 shunt (TIPS) in cirrhotic patients. Gut 1996; 39: 600–4
58 Borromeo CJ, Stix MS, Lally A, Pomfret EA. Epidural catheter and 80 Merli M, Valeriano V, Funaro S, et al. Modifications of cardiac func-
increased prothrombin time after right lobe hepatectomy for tion in cirrhotic patients treated with transjugular intrahepatic
living donor transplantation. Anesth Analg 2000; 91: 1139– 41 portosystemic shunt (TIPS). Am J Gastroenterol 2002; 97: 142– 8
59 Schumann R, Zabala L, Angelis M, Bonney I, Tighiouart H, Carr DB. 81 Iwakiri Y, Groszmann RJ. The hyperdynamic circulation of chronic
Altered hematologic profiles following donor right hepatectomy liver diseases: from the patient to the molecule. Hepatology 2006;
and implications for perioperative analgesic management. Liver 43: S121– 31
Transpl 2004; 10: 363 –8 82 Hennenberg M, Trebicka J, Sauerbruch T, Heller J. Mechanisms of
60 Siniscalchi A, Begliomini B, De Pietri L, et al. Increased prothrombin extrahepatic vasodilation in portal hypertension. Gut 2008; 57:
time and platelet counts in living donor right hepatectomy: impli- 1300–14
cations for epidural anesthesia. Liver Transpl 2004; 10: 1144– 9 83 Vaughan RB, Angus PW, Chin-Dusting JPF. Evidence for altered
61 Tsui SL, Yong BH, Ng KF, Yuen TS, Li CC, Chui KY. Delayed epidural vascular responses to exogenous endothelin-1 in patients with
catheter removal: the impact of postoperative coagulopathy. advanced cirrhosis with restoration of the norrmal vasoconstrictor
Anaesth Intensive Care 2004; 32: 630 –6 response following successful liver transplantation. Gut 2003; 52:
62 Hulshoff A, Schricker T, Elgendy H, Hatzakorzian R, Lattermann R. 1505–10
Albumin synthesis in surgical patients. Nutrition 2013; 29: 703– 7 84 Hall TH, Dhir A. Anesthesia for liver transplantation. Semin Cardio-
63 Vincent JL, Dubois MJ, Navickis RJ, Wilkes MM. Hypoalbuminemia thorac Vasc Anesth 2013; 17: 180–94
in acute illness: is there a rationale for intervention? A 85 Moller S, Hove JD, Dixen U, Bendtsen F. New insights into cirrhotic
meta-analysis of cohort studies and controlled trials. Ann Surg cardiomyopathy. Int J Cardiol 2013; 167: 1101–8
2003; 237: 319– 34 86 Zardi EM, Abbate A, Zardi DM, et al. Cirrhotic cardiomyopathy. J Am
64 Giannini EG, Testa R, Savarino V. Liver enzyme alteration: a guide Coll Cardiol 2010; 56: 539– 49
for clinicians. Can Med Assoc J 2005; 172: 367– 79 87 Biancofiore G, Mandell MS, Rocca GD. Perioperative considerations
65 Limdi JK, Hyde GM. Evaluation of abnormal liver function tests. in patients with cirrhotic cardiomyopathy. Curr Opin Anaesthesiol
Postgrad Med J 2003; 79: 307– 12 2010; 23: 128– 32
66 Hoekstra LT, de Graaf W, Nibourg GA, et al. Physiological and bio- 88 Giusca S, Jinga M, Jurcut C, Jurcut R, Serban M, Ginghina C. Porto-
chemical basis of clinical liver function tests: a review. Ann Surg pulmonary hypertension: from diagnosis to treatment. Eur J
2013; 257: 27– 36 Intern Med 2011; 22: 441– 7
67 Rochling FA. Evaluation of abnormal liver tests. Clin Cornerstone 89 Stauber RE, Olschewski H. Portopulmonary hypertension: short
2001; 3: 1–12 review. Eur J Gastroenterol Hepatol 2010; 22: 385– 90
68 Dib N, Oberti F, Cales P. Current management of the complications 90 Porres-Aguilar M, Altamirano JT, Torre-Delgadillo A, Charlton MR,
of portal hypertension: variceal bleeding and ascites. Can Med Duarte-Rojo A. Portopulmonary hypertension and hepatopul-
Assoc J 2006; 174: 1433–43 monary syndrome: a clinician-oriented overview. Eur Respir Rev
69 Berzigotti A, Seijo S, Reverter E, Bosch J. Assessing portal hyperten- 2012; 21: 223– 33
sion in liver disease. Expert Rev Gastroenterol Hepatol 2013; 7: 91 Kawut SM, Krowka MJ, Trotter JF, et al. Clinical risk factors for por-
141– 55 topulmonary hypertension. Hepatology 2008; 48: 196–203
70 Shibayama Y, Nakata K. Localization of increased hepatic vascular 92 Chen HS, Xing SR, Xu WG, et al. Portopulmonary hypertension in cir-
resistance in liver cirrhosis. Hepatology 1985; 5: 643 –8 rhotic patients: prevalence, clinical features and risk factors. Exp
71 Groszmann RJ, Abraldes JG. Portal hypertension—from bench to Ther Med 2013; 5: 819–24
bedside. J Clin Gastroenterol 2005; 39: S125–30 93 Krowka MJ, Plevak DJ, Findlay JY, Rosen CB, Wiesner RH, Krom RA.
72 Wiklund RA. Preoperative preparation of patients with advanced Pulmonary hemodynamics and perioperative cardiopulmonary-
liver disease. Crit Care Med 2004; 32: S106 –15 related mortality in patients with portopulmonary hypertension
73 Garcia-Tsao G, Bosche J. Management of varices and variceal undergoing liver transplantation. Liver Transpl 2000; 6: 443– 50
hemorrhage in cirrhosis. N Engl J Med 2010; 362: 823–32 94 Murray KF, Carithers RL Jr, AASLD. AASLD practice guidelines:
74 Villanueva C, Colomo A, Bosch A, et al. Transfusion strategies for evaluation of the patient for liver transplantation. Hepatology
acute upper gastrointestinal bleeding. N Engl J Med 2013; 368: 2005; 41: 1407– 32
11– 21 95 Ramsay MA, Simpson BR, Nguyen AT, Ramsay KJ, East C,
75 Gines P, Tito L, Arroyo V, et al. Randomized comparative study of Klintmalm GB. Severe pulmonary hypertension in liver transplant
therapeutic paracentesis with and without intravenous albumin candidates. Liver Transpl 1997; 3: 494– 500
in cirrhosis. Gastroenterology 1988; 94: 1493–502 96 Marik PE, Baram M, Vahid B. Does central venous pressure predict
76 Boyer TD, Haskal ZJ, American Association for the Study of Liver fluid responsiveness? A systematic review of the literature and the
Diseases. The role of transjugular intrahepatic portosystemic tale of seven mares. Chest 2008; 134: 172–8

i59
BJA Kiamanesh et al.

97 Burger-Klepp U, Karatosic R, Thum M, et al. Transesophageal echo- 119 Quittnat F, Gross P. Vaptans and the treatment of water-retaining
cardiography during orthotopic liver transplantation in patients disorders. Semin Nephrol 2006; 26: 234– 43
with esophagoastric varices. Transplantation 2012; 94: 192– 6 120 Ayus JC, Krothapalli RK, Arieff AI. Treatment of symptomatic hypo-
98 Suriani RJ, Cutrone A, Feierman D, Konstadt S. Intraoperative natremia and its relation to brain damage. A prospective study.
transesophageal echocardiography during liver transplantation. N Engl J Med 1987; 317: 1190– 5
J Cardiothorac Vasc Anesth 1996; 10: 699 –707 121 Hantman D, Rossier B, Zohlman R, Schrier R. Rapid correction of
99 Hemprich U, Papadakos PJ, Lachmann B. Respiratory failure and hyponatremia in the syndrome of inappropriate secretion of anti-
hypoxemia in the cirrhotic patient including hepatopulmonary diuretic hormone. An alternative treatment to hypertonic saline.
syndrome. Curr Opin Anaesthesiol 2010; 23: 133–8 Ann Intern Med 1973; 78: 870–5
100 Fallon MB, Abrams GA. Pulmonary dysfunction in chronic liver 122 Sterns RH, Riggs JE, Schochet SS Jr. Osmotic demyelination syn-
disease. Hepatology 2000; 32: 859– 65 drome following correction of hyponatremia. N Engl J Med 1986;
101 Bernal W, Auzinger G, Dhawan A, Wendon J. Acute liver failure. 314: 1535–42
Lancet 2010; 376: 190 –210 123 Nordlie RC, Foster JD, Lange AJ. Regulation of glucose production
102 Rodriguez-Roisin R, Krowka MJ, Herve P, Fallon MB, Committee by the liver. Annu Rev Nutr 1999; 19: 379– 406
ERSTFP-HVDS. Pulmonary-hepatic vascular disorders (PHD). Eur 124 Del Vecchio Blanco C, Gentile S, Marmo R, Carbone L, Coltori M.
Respir J 2004; 24: 861 –80 Alterations of glucose metabolism in chronic liver disease.
103 Rodriguez-Roisin R, Krowka MJ. Hepatopulmonary syndrome—a Diabetes Res Clin Pract 1990; 8: 29– 36
liver-induced lung vascular disorder. N Engl J Med 2008; 358: 125 Takikawa Y, Harada M, Wang T, Suzuki K. Usefulness and accur-
2378– 87 acy of the international normalized ratio and activity percent
104 Zhang J, Fallon MB. Hepatopulmonary syndrome: update on of prothrombin time in patients with liver disease. Hepatol Res
pathogenesis and clinical features. Nat Rev Gastroenterol 2013 Advance Access published on 21 February 2013. doi:
Hepatol 2012; 9: 539– 49 10.1111/hepr.12093
105 Tanikella R, Fallon MB. Hepatopulmonary syndrome and liver 126 Tripodi A, Mannucci PM. The coagulopathy of chronic liver disease.
transplantation: who, when and where? Hepatology 2013; 57: N Engl J Med 2011; 365: 147– 56
2097– 9 127 Northup PG, Caldwell SH. Coagulation in liver disease: a guide for
106 Krowka MJ, Mandell MS, Ramsay MA, et al. Hepatopulmonary syn- the clinician. Clin Gastroenterol 2013; 11: 1064–74
drome and portopulmonary hypertension: a report of the multi- 128 Tripodi A, Caldwell SH, Hoffman M, Trotter JF, Sanyal AJ. Review
center liver transplant database. Liver Transpl 2004; 10: 174– 82 article: the prothrombin time test as a measure of bleeding risk
107 Hartleb M, Gutkowski K. Kidneys in chronic liver diseases. World J and prognosis in liver disease. Aliment Pharmacol Ther 2007; 26:
Gastroenterol 2012; 18: 3035– 49 141– 8
108 Meltzer J, Brentjens TE. Renal failure in patients with cirrhosis: 129 Dalmau A, Sabate A, Aparicio I. Hemostasis and coagulation mon-
hepatorenal syndrome and renal support strategies. Curr Opin itoring and management during liver transplantation. Curr Opin
Anaesthesiol 2010; 23: 139 –44 Organ Transplant 2009; 14: 286–90
109 Gines P, Schrier RW. Renal failure in cirrhosis. N Engl J Med 2009; 130 Krzanicki D, Sugavanam A, Mallett S. Intra-operative hypercoagul-
361: 1279–90 ability during liver transplantation as demonstrated by thrombo-
110 Steadman RH, Van Rensburg A, Kramer DJ. Transplantation for elastography. Liver Transpl 2013; 19: 852–61
acute liver failure: perioperative management. Curr Opin Organ 131 Findlay JY, Rettke SR. Poor prediction of blood transfusion require-
Transplant 2010; 15: 368 –73 ments in adult liver transplantations from preoperative variables.
111 Anderson CL, Saad WE, Kalagher SD, et al. Effect of transjugular J Clin Anesth 2000; 12: 319–23
intrahepatic portosystemic shunt placement on renal function: 132 Massicotte L, Beaulieu D, Thibeault L, et al. Coagulation defects do
a 7-year, single-center experience. J Vasc Interv Radiol 2010; 21: not predict blood product requirements during liver transplant-
1370– 6 ation. Transplantation 2008; 85: 956– 62
112 Ginès P, Guevara M, Arroyo V, Rodés J. Hepatorenal syndrome. 133 Steib A, Freys G, Lehmann C, Meyer C, Mahoudeau G. Intraopera-
Lancet 2003; 362: 1819–27 tive blood losses and transfusion requirements during adult liver
113 Salerno F, Gerbes A, Gines P, Wong F, Arroyo V. Diagnosis, preven- transplantation remain difficult to predict. Can J Anaesth 2001;
tion and treatment of hepatorenal syndrome in cirrhosis. Postgrad 48: 1075–9
Med J 2008; 84: 662 –70 134 Lisman T, Caldwell SH, Burroughs AK, et al. Hemostasis and throm-
114 Gluud LL, Christensen K, Christensen E, Krag A. Terlipressin for bosis in patients with liver disease: the ups and downs. J Hepatol
hepatorenal syndrome. Cochrane Database Syst Rev 2012; 9: 2010; 53: 362– 71
CD005162 135 Martinez J, MacDonald KA, Palascak JE. The role of sialic acid in
115 Arroyo V, Terra C, Gines P. New treatments of hepatorenal syn- the dysfibrinogenemia associated with liver disease: distribution of
drome. Semin Liver Dis 2006; 26: 254– 64 sialic acid on the constituent chains. Blood 1983; 61: 1196–202
116 Gonwa TA, Morris CA, Goldstein RM, Husberg BS, Klintmalm GB. 136 Caldwell SH, Hoffman M, Lisman T, et al. Coagulation disorders
Long-term survival and renal function following liver transplant- and hemostasis in liver disease: pathophysiology and critical as-
ation in patients with and without hepatorenal syndrome—ex- sessment of current management. Hepatology 2006; 44: 1039–46
perience in 300 patients. Transplantation 1991; 51: 428–30 137 Colucci M, Binetti BM, Branca MG, et al. Deficiency of thrombin acti-
117 Seu P, Wilkinson AH, Shaked A, Busuttil RW. The hepatorenal vatable fibrinolysis inhibitor in cirrhosis is associated with
syndrome in liver transplant recipients. Am Surg 1991; 57: increased plasma fibrinolysis. Hepatology 2003; 38: 230–7
806– 9 138 Lisman T, Leebeek FW, Mosnier LO, et al. Thrombin-activatable
118 Gines P, Guevara M. Hyponatremia in cirrhosis: pathogenesis, fibrinolysis inhibitor deficiency in cirrhosis is not associated
clinical significance, and management. Hepatology 2008; 48: with increased plasma fibrinolysis. Gastroenterology 2001; 121:
1002– 10 131– 9

i60
Monitoring and managing hepatic disease in anaesthesia BJA
139 Lisman T, Leebeek FW. Hemostatic alterations in liver disease: a in fulminant hepatic failure. Curr Opin Anaesthesiol 2010; 23:
review on pathophysiology, clinical consequences, and treat- 121–7
ment. Dig Surg 2007; 24: 250 –8 153 Bajaj JS, Pinkerton SD, Sanyal AJ, Heuman DM. Diagnosis and
140 Mallett SV, Chowdary P, Burroughs AK. Clinical utility of viscoelastic treatment of minimal hepatic encephalopathy to prevent motor
tests of coagulation in patients with liver disease. Liver Int 2013; vehicle accidents: a cost-effectiveness analysis. Hepatology
33: 961– 74 2012; 55: 1164– 71
141 Tripodi A, Primignani M, Chantarangkul V, et al. The coagulopathy 154 Riordan SM, Williams R. Treatment of hepatic encephalopathy.
of cirrhosis assessed by thromboelastometry and its correlation N Engl J Med 1997; 337: 473– 9
with conventional coagulation parameters. Thromb Res 2009; 155 Bjerring PN, Eefsen M, Hansen BA, Larsen FS. The brain in acute liver
124: 132–6 failure. A tortuous path from hyperammonemia to cerebral
142 Pham HP, Shaz BH. Update on massive transfusion. Br J Anaesth edema. Metab Brain Dis 2009; 24: 5– 14
2013; 111 (Suppl. 1): i71– i82 156 Bass N, Mullen K, Sanyal A, et al. Rifaximin treatment in hepatic en-
143 Sihler KC, Napolitano LM. Complications of massive transfusion. cephalopathy. N Engl J Med 2010; 362: 1071– 81
Chest 2010; 137: 209 –20 157 Sakamoto M, Perry W, Hilsabeck RC, Barakat F, Hassanein T.
144 Friederich PW, Henny CP, Messelink EJ, et al. Effect of recombinant Assessment and usefulness of clinical scales for semiquantification
activated factor VII on perioperative blood loss in patients under- of overt hepatic encephalopathy. Clin Liver Dis 2012; 16: 27–42
going retropubic prostatectomy: a double-blind placebo- 158 Blei AT, Cordoba J. Hepatic encephalopathy. Am J Gastroenterol
controlled randomised trial. Lancet 2003; 361: 201 –5 2001; 96: 1968– 76
145 Lodge JP, Jonas S, Jones RM, et al. Efficacy and safety of repeated 159 Ong JP, Aggarwal A, Krieger D, et al. Correlation between ammonia
perioperative doses of recombinant factor VIIa in liver transplant- levels and the severity of hepatic encephalopathy. Am J Med 2003;
ation. Liver Transpl 2005; 11: 973– 9 114: 188–93
146 Bernstein DE, Jeffers L, Erhardtsen E, et al. Recombinant factor 160 Nielsen HB, Tofteng F, Wang LP, Larsen FS. Cerebral oxygenation
VIIa corrects prothrombin time in cirrhotic patients: a preliminary determined by near-infrared spectrophotometry in patients
study. Gastroenterology 1997; 113: 1930–7 with fulminant hepatic failure. J Hepatol 2003; 38: 188– 92
147 Jeffers L, Chalasani N, Balart L, Pyrsopoulos N, Erhardtsen E. Safety 161 Tofteng F, Jorgensen L, Hansen BA, Ott P, Kondrup J, Larsen FS.
and efficacy of recombinant factor VIIa in patients with liver Cerebral microdialysis in patients with fulminant hepatic failure.
disease undergoing laparoscopic liver biopsy. Gastroenterology Hepatology 2002; 36: 1333– 40
2002; 123: 118– 26 162 Jalan R, O Damink SW, Deutz NE, Lee A, Hayes PC. Moderate hypo-
148 Levy JH, Fingerhut A, Brott T, Langbakke IH, Erhardtsen E, Porte RJ. thermia for uncontrolled intracranial hypertension in acute liver
Recombinant factor VIIa in patients with coagulopathysecondary failure. Lancet 1999; 354: 1164– 8
to anticoagulant therapy, cirrhosis, or severe traumatic injury: 163 Jalan R, Olde Damink SW, Deutz NE, et al. Moderate hypothermia
review of safety profile. Transfusion 2006; 46: 919 –33 prevents cerebral hyperemia and increase in intracranial pressure
149 Shao YF, Yang JM, Chau GY, et al. Safety and hemostatic effect of in patients undergoing liver transplantation for acute liver failure.
recombinant activated factor VII in cirrhotic patients undergoing Transplantation 2003; 75: 2034– 9
partial hepatectomy: a multicenter, randomized, double-blind, 164 Jalan R, Olde Damink SW, Deutz NE, Hayes PC, Lee A. Moderate
placebo-controlled trial. Am J Surg 2006; 191: 245– 9 hypothermia in patients with acute liver failure and
150 Cohen B, Matot I. Aged Erythrocytesa fine wine or sour grapes?. uncontrolled intracranial hypertension. Gastroenterology 2004;
Br J Anaesth 2013; 111 (Suppl. 1): i62– i70 127: 1338–46
151 Teperman LW, Peyregne VP. Considerations on the impact of 165 Dmello D, Cruz-Flores S, Matuschak GM. Moderate hypothermia
hepatic encephalopathy treatments in the pretransplant with intracranial pressure monitoring as a therapeutic paradigm
setting. Transplantation 2010; 89: 771–8 for the management of acute liver failure: a systematic review. In-
152 Zafirova Z, O’Connor M. Hepatic encephalopathy: current man- tensive Care Med 2010; 36: 210–3
agement strategies and treatment, including management 166 Stravitz RT, Larsen FS. Therapeutic hypothermia for acute liver
and monitoring of cerebral edema and intracranial hypertension failure. Crit Care Med 2009; 37: S258–64

Handling editor: H. C. Hemmings

i61

You might also like