Sei sulla pagina 1di 12

British Journal of Anaesthesia 111 (S1): i50i61 (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

Editors key points


Hepatic disease can lead to multisystem organ
dysfunction with multiple significant anaesthetic
implications.
Pathophysiological alterations can occur in
pharmacological responses, the systemic and pulmonary
circulations, coagulation, and neurological function.

The liver metabolizes drugs, food, and toxins; synthesizes procoagulants and anticoagulants; and regulates temperature,
glucose, and metabolism. When patients with liver disease
need surgery, anaesthesiologists face several perioperative
challenges. Patients with liver disease have physiological perturbations that range from mild hyperbilirubinemia of no clinical consequence to severe coagulopathy and metabolic
disarray (Fig. 1). This article reviews the current body of knowledge of perioperative assessment, monitoring, and management of hepatic disease in patients who will undergo surgery.

Perioperative risk assessment


Patients with liver disease who undergo non-hepatic surgery
have greater transfusion requirements, high infection risk,
cardiac compromise, longer hospital stays, and increased operative mortality.1 6 Patient-specific risk factors, various scoring
systems, and surgical procedures stratify risk for these
patients.6 9 Independent risk factors for complications and
mortality are outlined in Table 1.
The Child Turcotte Pugh (CTP) classification system, originally devised in 1964 to predict outcomes in cirrhotic patients
undergoing portosystemic surgery, assesses perioperative risk
for patients with liver disease who undergo hepatic or nonhepatic surgery.8 10 Scores are calculated from five variables:
serum albumin, serum bilirubin, ascites, prothrombin time
(PT), or INR (modified CTP score), and grade of encephalopathy
to stratify patients into three groups (Class Ascore 56; Class
Bscore 79; Class Cscore 1015).11 This scoring system
relies on a subjective assessment of ascites and encephalopathy and does not consider preoperative infection, aetiology
of cirrhosis, or surgery type.6 Original studies suggested high
mortality rates in Class A patients (10%), Class B patients

Keywords: liver; liver, cirrhosis; liver, disease; liver, function; liver,


metabolism

(82%), and Class C patients (82%) who undergo abdominal surgeries.12 13 A newer study reports lower mortality rates after
abdominal operations: Class A2%; Class B12%; Class
C12%.14 Improvements in surgical technique, anaesthetic
management, and perioperative care might explain the difference in these findings.
The model for end-stage liver disease (MELD) score is a validated and prognostic tool that estimates disease severity and
survival in patients with liver disease. The MELD score is objective, 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
number of liver diseases (alcoholic hepatitis and variceal
bleeding)16 and surgeries (abdominal, orthopaedic, and cardiovascular).5 8 17 19 In patients who have undergone abdominal surgery, an elevated MELD score was a better predictor of
poor perioperative outcome than CTP classification.17 Patients
with MELD scores .15 should avoid elective surgery.20 In
2002, the United Network for Organ Sharing adopted the
MELD score as a method of allocation of organs to estimate survival. The incorporation of hyponatraemia, an important prognostic factor in patients with cirrhosis, into the MELD score
might improve risk stratification among patients awaiting
liver transplantation.21
The type of surgical procedure influences perioperative outcomes in patients with hepatic disease. During abdominal
surgery, reflexive systemic hypotension from visceral traction
and blood loss can reduce hepatic arterial blood flow.22 23
Patients with extensive liver disease who undergo abdominal
surgery have a mortality rate of 30%.12 24 Mortality rates in
cirrhotic patients who undergo open cholecystectomy are
2350%; cardiac surgery, 16 31%; laparotomy for trauma,
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

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

Perioperative risk is determined by both patient-specific


and procedural factors.

Summary. Patients with liver disease have multisystem organ


dysfunction that leads to physiological perturbations ranging
from hyperbilirubinaemia of no clinical consequence to severe
coagulopathy and metabolic disarray. Patient-specific risk
factors, clinical scoring systems, and surgical procedures
stratify perioperative risk for these patients. The anaesthetic
management of patients with hepatic dysfunction involves
consideration of impaired drug metabolism, hyperdynamic
circulation, perioperative hypoxaemia, bleeding, thrombosis,
and hepatic encephalopathy.

BJA

Monitoring and managing hepatic disease in anaesthesia

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

Hepatic disease
Cardiovascular
Vasodilated state
Hyperdynamic cardiac function
Cirrhotic cardiomyopathy
Drug-induced hypertension
Portopulmonary hypertension

Decreased metabolic function


Decreased gluconeogenesis
Decreased lactate clearance
Hyperbilirubinemia
Varices
Portal hypertension
Ascites

Neurologic
Hepatic encephalopathy
Cerebral oedema
Intracranial hypertension

Pulmonary
Aspiration risk
Hepatopulmonary syndrome
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


and mortality1 33
Scoring system
Modified Child Turcotte Pugh (CTP)
Model for end-stage liver disease (MELD)
ASA-PS IV or V
Patient-specific risk factors

Male gender
Age .70 yr
Preoperative infection
Cirrhosis other than primary biliary cirrhosis
Elevated creatinine
Chronic obstructive pulmonary disease
Intraoperative hypotension
Upper gastrointestinal bleeding
Ascites

High-risk surgeries

Abdominal surgery
Open cholecystectomy
Cardiothoracic surgery
Trauma laparotomy
Oesophageal surgery
Emergency surgery

coagulopathy or CTP Class C cirrhosis are at even higher risk


during open cholecystectomies, abdominal surgeries, and
cardiovascular procedures.25 27 29 30 32 Contraindications to
elective surgery include fulminant hepatic failure, acute viral
or alcoholic hepatitis, chronic active symptomatic hepatitis,
CTP Class C cirrhosis, severe coagulopathy (platelet
count,50 000 ml21, uncorrectable PT .3 s above control),
hypoxaemia, heart failure, and acute kidney injury.3 34

Management of anaesthetic medications


and metabolism
The metabolic activity and clearance of anaesthetic medications is impaired in patients with liver disease. Without
adjusted doses of medications, duration of drug action is
extended and toxic levels can result.35 36 Emergence from anaesthesia can be delayed.
Hepatic metabolism takes place via two pathways: phase 1,
biotransformation via the cytochrome P450 system (which
requires delivery of oxygen via hepatic perfusion); and phase 2,
glucuronide conjugation. The first is decreased in liver disease;
the second is relatively spared when dysfunction begins.37

i51

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

Haematologic
Bleeding and thrombosis
Unreliable INR
Decreased factor synthesis
Thrombocytopenia and platelet dysfunction
Dysfibrinogenaemia

BJA

i52

can develop.58 60 Delayed epidural catheter removal has been


suggested in patients who undergo hepatectomy.61

Organ response, assessment, and


management
Hepatic response
Laboratory tests, such as albumin and bilirubin, are non-specific
and can be affected by other diseases and medications. Even
with hepatic disease, patients can have normal liver function
and laboratory values because the liver, the largest organ and
gland in the body, has a large reserve capacity.
Albumin, produced exclusively in the liver, has been used to
assess hepatic synthetic function.62 Low levels of this nonspecific marker are also found in patients with malnutrition,
renal disease, and capillary leak syndrome. This prognostic
marker in hospitalized patients is a negative acute phase reactant because the production of albumin decreases during states
of inflammation with cytokine release and surgical tissue
trauma.62 63 Albumin, with a long half-life of 20 days, can be
normal even in acute liver failure.64
The breakdown of red blood cells within the reticuloendothelial system forms unconjugated (indirect) bilirubin. Unconjugated bilirubin is transported to the liver to be conjugated
and secreted into bile.64 65 Increased production, decreased
hepatic uptake, and decreased conjugation of bilirubin
cause unconjugated hyperbilirubinaemia. The most common
causes of unconjugated hyperbilirubinaemia are haemolysis,
resolution of haematomas, inborn errors of bilirubin conjugation (Gilberts syndrome and Crigler Najjar syndrome), or
portal hypertension.64 Under normal circumstances, levels of
conjugated bilirubin in serum are negligible after rapid excretion into the biliary system. Conjugated hyperbilirubinaemia
can indicate intrinsic liver disease, especially in conjunction
with elevated aminotransferase levels, a sign of hepatocellular
injury. Conjugated hyperbilirubinaemia with elevated alkaline
phosphatase and minimal increase in aminotransferase
levels is consistent with a purely cholestatic process and can
result from numerous causes, including cholestatic drug reactions or obstructive jaundice.64 67
The normal portosystemic arterial pressure gradientbetween
the portal vein and hepatic vein is ,5 mm Hg. Intrahepatic vascular resistance and portal venous blood flow increase the gradient, characteristic of portal hypertension. Vascular resistance
in the liver can result from several factors: liver fibrosis, regenerative nodules, collagen deposits in the space of Disse,
endothelial dysfunction, imbalance between endogenous
vasoconstrictors (i.e. norepinephrine, thromboxane A, endothelin I, angiotensin II, and leukotrienes) and vasodilators (nitric
oxide), and contraction of portal and septal myofibroblasts
and smooth muscle cells.68 70 As the pressure gradient
between the portal vein and the hepatic vein increases, gastrooesophageal and haemorrhoidal varices develop to decompress
the portal system. Portal hypertension produces endogenous
vasodilators that cause splanchnic vasodilation.68 69 71 Splanchnic vasodilation in turn increases portal blood flow and exacerbates portal hypertension.

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

Many anaesthetic drugs (i.e. opioids, barbiturates) bind to


albumin in the circulation. Low albumin levels increase the
free fraction of drug and can theoretically exaggerate pharmacological effect.38 End-stage liver disease alters hepatic extraction, renal excretion, and levels of serum proteins in addition to
albumin. The net effect of these changes is unpredictable,
making general recommendations for drug administration difficult. For safety, drugs are titrated to desired effect.
Patients with hepatic dysfunction can be sensitive to sedatives such as midazolam and diazepam.39 40 In contrast, the
pharmacokinetics of lorazepam, which undergoes glucuronidation, is less likely to be affected by liver disease.41 Benzodiazepines can precipitate hepatic encephalopathy, and flumazenil
can reverse this condition.42 Conversely, patients who have a
history of alcohol abuse can require large doses of benzodiazepines. Rapid redistribution of bolused propofol determines its
duration of action, which is not affected by impaired liver metabolism. In a small study of cirrhotic patients, propofol infusions
did not increase termination half-life or clearance to a clinically
significant degree.43
The adverse effects of morphine can be more pronounced
in patients with severe liver disease.44 45 Morphine is metabolized by glucuronidation in the liver, and its intermediate
metabolites are eliminated renally. Although the pharmacokinetics of morphine can be unaltered in patients with mild
hepatic disease, a prolonged half-life is associated with a prolonged PT, hypoalbuminaemia, encephalopathy, ascites, and
jaundice.37 46 47 The elimination of morphine metabolites can
be prolonged in patients with hepatorenal syndrome (HRS).
Metabolism of fentanyl, sufentanil, and morphine, which
have a high hepatic extraction ratio, is slowed by impaired
hepatic blood flow.38 44 48
In patients with liver disease and oedema, neuromuscular
blocking agent doses must be augmented because fluid retention increases the volume of distribution. Neuromuscular
blocking agents that are metabolized by the liver, such as
vecuronium and rocuronium, have a slower onset and longer
duration of action.49 54 Liver disease reduces plasma cholinesterase activity, and prolonged neuromuscular block after
succinylcholine has been reported.55 Acute kidney injury with
elevated potassium levels or immobilization for long periods
might preclude the use of succinylcholine.
Vasodilating anaesthetic agents such as propofol and volatile anaesthetics cause hypotension if intravascular volume is
depleted (i.e. by overdiuresis or gastrointestinal losses), and
reduce liver perfusion. In addition to perioperative haemorrhage and hypotension, volatile anaesthetic agents can
reduce hepatic blood flow and hepatic oxygen uptake. Compared with the vasoconstrictive effects of halothane, isoflurane and desflurane can improve hepatic blood flow and
splanchnic perfusion via hepatic vasodilation.56 57
Patients with end-stage liver disease often exhibit coagulation disorders. The presence of a coagulopathy is a contraindication to regional anaesthesia, particularly epidural anaesthesia.
In patients without a preoperative disorder of coagulation,
intraoperative or postoperative coagulopathy from hepatic ischaemia, insufficient residual hepatic parenchyma, and dilution

Kiamanesh et al.

Monitoring and managing hepatic disease in anaesthesia

Cardiovascular response
The cardiovascular system of patients with end-stage liver
disease mimics the hyperdynamic circulatory changes in
patients with sepsis. Tachycardia, elevated cardiac output, low
arterial pressure, and low systemic vascular resistance are characteristic.81 Enhanced endogenous production or diminished
hepatic clearance of vasodilating substances (nitric oxide,
carbon monoxide, and endogenous cannabinoids, tumour necrosis factor-a, adrenomedullin, and hydrogen sulfide) and the
inflammatory response to bacterial translocation cause
splanchnic arterial vasodilation (Fig. 2).81 82 These changes
have been reversed with liver transplantation.81 83 In response

to splanchnic vasodilation and decreased systemic vascular resistance, the body retains sodium and water, which increases
plasma volume. An increase in cardiac output and heart rate
follow.81 84 Venous capacitance increases from formation of portosystemic shunts with portal hypertension to contribute to a
hyperdynamic circulation.
Management of vasodilatory hypotension focuses on
expanding the intravascular volume, administering a vasoconstricting agent such as norepinephrine or vasopressin, and determining the underlying cause of vasodilation to target.
Aggressive fluid resuscitation without assessment of fluid responsiveness should be avoided because excessive i.v. fluid administration to a non-fluid-responsive patient can increase
cardiac filling pressures, which can cause hepatic congestion,
pulmonary oedema, and resulting respiratory failure.
Systemic conditions such as haemochromatosis (ventricular
hypertrophy with increased end-diastolic and end-systolic
volumes), amyloidosis (restrictive cardiomyopathy), Wilsons
disease (supraventricular extrasytolic beats), and alcoholism
(systolic and diastolic dysfunction) can affect liver and cardiac
function.85 During the perioperative period, the circulatory
system can be challenged with shunt insertion or greater afterload from surgical stress, unmasking underlying cirrhotic cardiomyopathy. Echocardiography shows impaired myocardial
function similar to that found in sepsis.86 This condition is also
characterized by QT prolongation and systolic and diastolic dysfunction.87 Reduced b-adrenoceptor function might explain
these findings.85 Cirrhotic cardiomyopathy is often reversible
and requires inotropic and diuretic support to prevent elevation
of central venous pressure and hepatic congestion.
Patients with portal hypertension can develop portopulmonary hypertension (PPHTN) and right ventricular dysfunction.
While the exact pathogenesis of PPHTN remains unclear, histopathological findings are indistinguishable from those observed
in idiopathic pulmonary artery hypertension (medial hypertrophy, concentric intimal proliferation, plexiform lesions, in
situ thrombosis, and fibrotic changes).88 89 A hyperdynamic circulation might increase shear stress on the pulmonary vasculature to cause remodelling and pulmonary hypertension.88 90 The
mean pulmonary artery pressure .25 mm Hg at rest and pulmonary vascular resistance .240 dyn s cm25 define PPHTN.
PPHTN has been reported in 69% of patients with advanced
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
between 35 and 50 mm Hg and 100% in patients with pressures
.50 mm Hg.93 Calcium channel blockers, which can be used in
patients with pulmonary arterial hypertension, are not recommended in PPHTN because they produce mesenteric vasodilatation and worsen portal hypertension. b-Blocker therapy, which
is often used in patients with portal hypertension to reduce
the risk of variceal bleeding, is contraindicated in patients
with PPHTN because its negative inotropic effect decreases
exercise tolerance.88 90 Finally, while liver transplantation is a
tempting option to reduce PPHTN, moderate or severe uncontrolled PPHTN is associated with significant mortality risk and
is a contraindication to OLT.93 95 Intraoperative management

i53

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

Complications of portal hypertension include gastrointestinal


bleeding from expansion and rupture of varices, intraoperative
bleeding from intra-abdominal collaterals, and formation of
ascites and pleural effusions.68 72 Almost half of patients with cirrhosis have oesophageal varices at the time of diagnosis, with
the 1 yr risk of variceal haemorrhage at 12% and a 1 yr rate of recurrent haemorrhage at 60%. The 6 week mortality with each
episode of variceal haemorrhage is between 15% and 20%.73
In patients with acute upper gastrointestinal bleeding, a restrictive transfusion strategy (transfusion when the haemoglobin is
,7 g dl21) can decrease subsequent bleeding and prevent an increase in the portal-pressure gradient.74 Patients who have
received multiple previous transfusions, such as patients with a
history of gastrointestinal bleeding, should be evaluated in
advance for cross-matching large quantities of blood products
possibly required for a procedure.
Before operation, ascites is initially managed with a low
sodium diet and diuretic therapy. Patients with ascites refractory to treatment with diuretics require large-volume paracentesis and albumin replacement to prevent hypotension and
HRS.75 Large-volume abdominal ascites can lead to a lower
venous return and, in conjunction with pleural effusions, can
decrease pulmonary reserve and prevent patients from lying
flat. If there is evidence of tense ascites, recent food ingestion,
or a propensity for delayed gastric emptying, a rapid-sequence
induction of general anaesthesia with tracheal intubation
should be considered.
A transjugular intrahepatic portosystemic shunt (TIPS) prevents recurrent variceal bleeding and manages refractory
ascites.76 During the procedure, an expandable stent is placed
into the liver parenchyma for portosystemic exchange to decrease portal hypertension. The procedure is performed under
local or general anaesthesia as a bridge to liver transplantation
in patients with severe liver disease.77 General anaesthesia
might be preferred because of the duration of the procedure
and the potential for a life-threatening event such as bleeding,
infection, haemobilia, arrhythmias, liver ischaemia, and stent
migration.78 Preoperative evaluation of cardiac function (particularly right ventricular function) is essential because stent
placement can rapidly increase venous return and cardiac
output to unmask underlying cardiomyopathy.79 80 Encephalopathy can worsen from enhanced systemic delivery of biogenic
amines normally cleared by the liver.

BJA

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
Splanchnic vasodilation
Systemic vasodilation

Liver dysfunction
Portal hypertension
Bacterial translocation and inflammation

Fig 2 Vasodilation from hepatic dysfunction.

is targeted to prevent increases in pulmonary vascular resistance by controlling hypoxia, hypercarbia, acidosis, pain, and
hypothermia.
Because of these cardiovascular changes, invasive monitors
usually are used during the perioperative period. Beat-to-beat
arterial pressure is monitored and blood is sampled frequently
via a radial artery catheter. Pulse pressure variation, calculated
from the invasive arterial tracing, assesses fluid responsiveness. Peripheral arterial pressure measurements can be unreliable in patients with severe vasoconstriction or vasodilation.
Central venous access is established to measure right atrial
pressure and to administer drugs. Monitoring central venous
pressure does not reliably measure blood volume or change
in blood volume.96 Echocardiography assesses ventricular
filling, contractility, and function. Myocardial ischaemia, pulmonary embolism, pleural effusion, and inadequate inferior
vena cava reconstruction can be observed with transoesophageal echocardiography (TOE). The risk of rupture of oesophageal varices with TOE is rare.97 98 Bladder pressures should
be measured if intra-abdominal hypertension from ascites is
suspected.

Pulmonary response
Up to 70% of patients with end-stage liver disease complain of
dyspnoea.99 100 Potential causes of preoperative hypoxaemia
and respiratory failure in patients with cirrhosis include atelectasis from the compressive effects of ascites, hepatic
hydrothorax, hepatopulmonary syndrome (HPS), underlying

i54

chronic pulmonary disease and occasionally acute respiratory


distress syndrome.99 Chest radiography rules out pulmonary
oedema, pleural effusions, or cardiomegaly in patients with
symptoms of dyspnoea. Muscle wasting and intra-abdominal
hypertension from ascites increase the work of breathing.
Ascites fluid can enter the pleural space through small channels in the diaphragm to cause a hepatic hydrothorax (usually
on the right side). Compressive atelectasis from ascites and
pleural effusions reduce functional residual capacity and decrease the time to oxygen desaturation. Lung recruitment
and pleural fluid drainage can improve pulmonary mechanics.
Respiratory alkalosis can be observed in patients with a high
respiratory drive from liver dysfunction. The use of positivepressure mechanical ventilation and PEEP can lead to an
increased average intrathoracic pressure, which decreases
venous return from the inferior vena cava and hepatic veins
and can lead to congestion of the liver. This concern remains
controversial, as studies with PEEP of up to 10 cm H2O have
shown no adverse effects on hepatic arterial, portal venous, or
hepatic venous flow.101
A diagnosis of HPS is considered in patients without cardiopulmonary disease who have a PaO2 ,8.0 kPa. The triad of liver
disease and/or portal hypertension, widened age-corrected alveolararterial oxygen gradient (.22.7 kPa) while breathing
room air, and documented intrapulmonary vascular dilation
(IPVD) define HPS.99 102 103 Enhanced production or impaired
hepatic clearance of endogenous vasodilators (i.e. nitric oxide,
vasodilator prostaglandins, substance P), or inhibition of vasconstrictive substances (i.e. tyrosine, serotonin, and endothelin) by a

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

Increased plasma volume


Increased cardiac output

Monitoring and managing hepatic disease in anaesthesia

damaged livercan cause IPVD. IPVD causes hypoxaemiaviaventilation and perfusion mismatching, intrapulmonary shunt
physiology, and diffusion limitation.99 103 Patients with HPS
often complain of dyspnoea and fatigue; they can have clubbing
and spider angiomata. They may experience platypnoea (dyspnoea in the upright position relieved by recumbency) or have
orthodeoxia (arterial oxyhaemoglobin desaturation in the
upright position). Preferential perfusion of IPVDs with the
patient upright might cause these clinical manifestations.
Contrast-enhanced echocardiography or perfusion lung scanning with technetium-99m-labelled macroaggregate albumin
detects intrapulmonary shunting suggestive of IPVD.102 103
Resolution of this syndrome after transplant has been reported.
Medical therapy for HPS has, to date, been relatively ineffective.
Currently, the only known treatment option for patients with HPS
is liver transplantation.104 106

Renal, electrolyte, and glucose response

renal function is impaired rapidly and progressively with a doubling of the initial serum creatinine to .2.5 mg dl21 or a 50% reduction in the 24 h creatinine clearance to ,20 ml min21 in ,2
weeks. Patients with type II HRS have impaired renal function
and a serum creatinine .1.5 mg dl21 and do not meet the criteria for type I HRS.107 Compared with patients diagnosed with
prerenal failure, patients with HRS lack a renal response to a
1.5 litre volume challenge. Distinguishing between HRS and
acute tubular necrosis is difficult. Fractional excretion of
sodium ,1% suggests tubular function and favours a diagnosis
of HRS. The presence of renal tubular epithelial cells in urinary
sediment favours a diagnosis of acute tubular necrosis.109 112
Administration of terlipressin, a vasopressin analogue, along
with i.v. albumin has been shown to have some benefit in
patients with type 1 HRS.114 115 Medical management of HRS
is marginally effective, but liver transplantation usually reverses
HRS.116 117
Hypervolaemic hyponatraemia from accumulated secretion
of anti-diuretic hormone acts on vasopressin-2 receptors of
the renal tubular collecting duct to impair excretion of solutefree water.118 When extracellular volume expands, ascites and
oedema can result. In patients with hypovolaemic hyponatraemia via loss of extracellular fluid from the kidneys (overdiuresis)
or gastrointestinal tract, ascites or oedema is rare. These
patients can have prerenal failure from low circulating volume
or hepatic encephalopathy from rapid reduction in serum
osmolality.109 118 Distinguishing between hypervolaemic and
hypovolaemic hyponatremia guides treatment. Hypervolaemic
hyponatraemia is managed with fluid restriction (11.5 litre
day21) and withholding of diuretics. Vaptans, medications
that block the V2 receptor, increase solute-free water excretion
dose-dependently and might preclude water restriction so that
diuretics can be continued.119 Patients with hypovolaemic hyponatraemia are not given diuretics; instead, saline is administered
to increase plasma volume.118
Patients with preoperative hyponatraemia are at risk for
central pontine myelinolysis (CPM) perioperatively. CPM is a
neurological condition characterized by symmetric noninflammatory demyelinating lesions in the basis pontis. The aetiology of CPM is uncertain, but osmotic stress on central nervous
system cells has been suggested, and CPM correlates with rapid
correction of hyponatraemia.118 Rapid changes in concentration
of serum sodium cannot be predicted and a safe rate of correction of hyponatraemia has not been definitively established;
some experts recommend sodium correction at a rate ,12 mEq
litre21 day21.120 122 Preoperative correction of hyponatraemia
can prevent a rapid increase in serum sodium levels intraoperatively and after operation. Sodium bicarbonate should be administered cautiously because it has a high concentration of
sodium.
Calcineurin inhibitors, loop diuretics, and significant blood
loss can worsen hypomagnesaemia. Total serum calcium is
often low because patients commonly have low serum
albumin with reduced calcium binding. Failure to clear citrate
(metabolized by the liver) after the administration of a significant volume of packed red blood cells, malnutrition, and
vitamin D deficiency can lead to perioperative hypocalcaemia.

i55

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

Acute kidney injury commonly occurs in patients with chronic


liver disease and is present in up to 20% of patients hospitalized
with decompensated liver cirrhosis.107 Patients with acute
kidney injury and cirrhosis have more complications and
greater risk for mortality after liver transplantation than those
without renal failure. Gastrointestinal bleeding, diarrhoea from
infection or lactulose administration, and diuretic medications
change circulatory function via hypovolaemia and can cause prerenal injury. This injury rapidly reverses with correction of the
underlying pathophysiology and volume resuscitation.108 109
As cirrhosis progresses, reduction in systemic vascular resistance activates the reninangiotensin and sympathetic nervous
systems, which leads to ascites, oedema, and vasoconstriction of the intrarenal circulation and renal hypoperfusion.
Ischaemic events and medications such as non-steroidal antiinflammatory drugs or aminoglycosides cause intrinsic renal
damage.108 109 Managing kidney injury, low urine output, and
electrolyte abnormalities in patients with hepatic disease is
challenging. Pre- or post-renal kidney injuries reverse with
enhanced renal perfusion or removal of the obstruction, respectively. Optimizing renal perfusion and avoidance of
nephrotoxic drugs are also important in these patients.110 Continuous renal replacement therapy can be used perioperatively
to manage volume shifts, acid base balance, and electrolyte
disturbances. Placement of a TIPS can improve serum creatinine, with more profound effects on those patients with higher
baseline levels.111
HRS is caused by functional renal vasoconstriction (via activation of the reninangiotensin system, the sympathetic nervous
system, and arginine vasopressin) in response to splanchnic arterial vasodilation.109 112 Although histological findings and
diagnostic tests for HRS are lacking, diagnostic criteria are
used to categorize two types of HRS. Criteria for HRS include (i)
cirrhosis with ascites, (ii) serum creatinine .1.5 mg dl21, (iii)
no improvement of creatinine after 2 days of diuretic withdrawal
and volume expansion with albumin, (iv) absence of shock, (v)
no current or recent treatment with nephrotoxic drugs, and
(vi) absence of parenchymal kidney disease.113 In type I HRS,

BJA

BJA
Disorders of glucose metabolism are observed in patients
with chronic liver disease and cirrhosis.123 124 Multiple factors
affect glucose levels, including the inflammatory response to
surgery, steroid administration, hepatic dysfunction, altered
glycogen stores, and insulin resistance in liver failure. Episodes
of hypoglycaemia are observed in patients with end-stage liver
disease.

Haematological response

i56

thromboelastography or rotational thromboelastometry


assess the viscoelastic properties of a whole blood sample as a
function of time to reflect the function and interaction of
plasma, blood cells, and platelets. These measurements
detect a hypercoagulable state and distinguish hyperfibrinolysis
from other causes of coagulopathy, such as factor depletion or
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
are no studies evaluating these tests as predictors of intraoperative bleeding in patients with hepatic disease.129 130 140
Uncontrolled haemorrhage and massive transfusion in the
operating theatre or the intensive care unit can cause the
lethal triad of acidosis, coagulopathy, and hypothermia. Left
uncorrected, each of these abnormalities can exacerbate the
others, creating a bloody vicious cycle. Additional early complications of massive transfusion include (i) acute haemolytic
transfusion reactions, (ii) febrile non-haemolytic transfusion
reactions, (iii) transfusion-related acute lung injury (TRALI),
(iv) transfusion-associated circulatory overload, (v) allergic
reactions, (vi) bacterial sepsis, (vii) hypocalcaemia, and (viii)
hyperkalaemia (see Pham and Shaz,142 this issue).143
Warming the room, applying forced warming systems,
administering blood products and fluids through a fluid
warmer, and heating and humidifying inspired gases reduce
the risk of hypothermia. Dilutional coagulopathy and thrombocytopenia may be prevented by transfusing packed red blood
cells, fresh-frozen plasma, and platelets in a 1:1:1 ratio.143 Occasionally, recombinant Factor VIIa is administered.144 148 Factor
VIIa has not been shown to decrease red blood cell transfusion
requirements during hepatectomies.149 Potassium, magnesium, and calcium levels are frequently monitored to correct abnormal levels. Improving haemodynamic parameters, adjusting
minute ventilation, or administering sodium bicarbonate or tromethamine manage metabolic acidosis. The riskof TRALI can be
reduced by minimizing transfusions and transfusing packed red
blood cells with a short storage time (see Cohen and Matot,150
this issue) and fresh-frozen plasma from men or nulliparous
women.

Neurological response
Patients with liver disease can have encephalopathy before operation or after operation. Brain dysfunction manifests in acute
or chronic liver failure or after a spontaneous or surgical portosystemic shunt.151 152 Hepatic encephalopathy is a neuropsychiatric complication of acute and chronic liver disease
with features that range from mild confusion to cerebral
oedema with intracranial hypertension.101 152 Patients have
disturbances in consciousness, cognitive abilities, behaviour,
fine motor skills, concentration, reaction time, memory,
and/or electroencephalogram.152 154 The pathogenesis of
hepatic encephalopathy is not understood, but most theories
implicate elevated levels of ammonia, a gut-derived neurotoxin, which is shunted to the systemic circulation from the
portal system. Bacteria in the intestinal tract produce
ammonia, which crosses the blood brain barrier into

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

In patients with hepatic disease, haemostatic changes


promote both bleeding and thrombosis. Inadequate synthesis
of all coagulation factors (except for von Willebrands factor
and factor VIII), thrombocytopenia, platelet function defects,
dysfibrinogenaemia, and elevated tissue plasminogen activator (tPA) levels cause bleeding. Elevation of von Willebrands
factor and Factor VIII and decreased levels of ADAMTS-13 (a
disintegrin and metalloproteinase with a thrombospondin
type 1 motif, member 13), protein C, protein S, antithrombin,
a 2-macroglobulin, plasminogen, and heparin cofactor II
favour thrombosis.
Factors VII, X, V, II (prothrombin), and I (fibrinogen) have a
short half-life (hours to days) and are synthesized solely by hepatocytes, making possible a semi real-time evaluation of hepatic
synthetic function.125 Although patients with liver disease can
have abnormal PT from decreased production of pro-coagulant
factors, the rebalanced state of pro- and anti-coagulants in
patients with liver disease makes the PT an unreliable tool for
evaluating tendency to bleed or clot.126 133
Levels of fibrinogen, an acute phase reactant, are normal or
increased in mild-to-moderate liver disease. In patients with
severe hepatic dysfunction, however, fibrinogen is poorly
synthesized and dysfunctional, which increases the risk of
bleeding.134 135 Although hyperfibrinolysis (from increased
levels of tPA and reduced levels of thrombin-activatable fibrinolysis inhibitor and plasmin inhibitor) has been observed in cirrhotic patients, its association with bleeding is unclear.126 136
Hypofibrinolysis (from increased levels of plasminogen activator inhibitor and reduced levels of plasminogen) can restore the
balance of fibrinolysis.126 137 138
Thrombocytopenia and platelet dysfunction are characteristic of end-stage liver disease. Thrombocytopenia results
from several factors: portal hypertension with hypersplenism,
which sequesters platelets; consumption of platelets during
systemic intravascular coagulation; and impaired hepatic synthesis of thrombopoietin, which stimulates platelet production
in the bone marrow.72 139 Defective signal transduction;
uraemia from acute kidney injury; and intrinsic defects of
ADP, arachidonic acid, collagen, and thrombin prevent platelet
aggregation.139
Patients with active sites of bleeding should be transfused
with fresh-frozen plasma, packed red blood cells, cryoprecipitate, or platelets as dictated by clinical assessment and laboratory tests. In addition, desmopressin can be considered for
patients with concomitant renal dysfunction and uraemic
bleeding. Coagulation status should always be monitored by
inspection of the surgical field. Measurements from

Kiamanesh et al.

BJA

Monitoring and managing hepatic disease in anaesthesia

Conclusions
Patients with hepatic disease are at high risk for poor perioperative outcomes. Assessment of the severity of liver dysfunction
and consideration of type of surgery stratifies risk and delays
elective surgery in patients with significant hepatic disease
(CTP Class C cirrhosis). Managing surgical patients with hepatic
disease considers anaesthetic dose adjustment and monitoring
the multisystem organ responses to liver dysfunction.

Authors contributions
All authors participated in literature search, writing of first
draft, editing, and final approval.

Acknowledgement
The authors would like to thank Sally Kozlik for her invaluable
editorial assistance.

Declaration of interest
None declared.

References
1

Csikesz NG, Nguyen LN, Tseng JF, Shah SA. Nationwide volume and
mortality after elective surgery in cirrhotic patients. J Am Coll Surg
2009; 208: 96 103
2 del Olmo JA, Flor-Lorente B, Flor-Civera B, et al. Risk factors for nonhepatic surgery in patients with cirrhosis. World J Surg 2003; 27:
64752
3 Friedman LS. The risk of surgery in patients with liver disease.
Hepatology 1999; 29: 161723
4 Rice HE, OKeefe GE, Helton WS, Johansen K. Morbid prognostic
features in patients with chronic liver failure undergoing nonhepatic surgery. Arch Surg 1997; 132: 8804; discussion 4 5
5 Teh SH, Nagorney DM, Stevens SR, et al. Risk factors for mortality
after surgery in patients with cirrhosis. Gastroenterology 2007;
132: 1261 9
6 Ziser A, Plevak DJ, Wiesner RH, Rakela J, Offord KP, Brown DL. Morbidity and mortality in cirrhotic patients undergoing anesthesia
and surgery. Anesthesiology 1999; 90: 4253
7 Cho HC, Jung HY, Sinn DH, et al. Mortality after surgery in patients
with liver cirrhosis: comparison of Child Turcotte Pugh, MELD
and MELDNa score. Eur J Gastroenterol Hepatol 2011; 23: 51 9
8 Farnsworth N, Fagan SP, Berger DH, Awad SS. Child TurcottePugh
versus MELD score as a predictor of outcome after elective and
emergent surgery in cirrhotic patients. Am J Surg 2004; 188:
5803
9 Hoteit MA, Ghazale AH, Bain AJ, et al. Model for end-stage liver
disease score versus child score in predicting the outcome of surgical procedures in patients with cirrhosis. World J Gastroenterol
2008; 14: 177480
10 Cholongitas E, Marelli L, Shusang V, et al. A systematic review of
the performance of the model for end-stage liver disease (MELD)
in the setting of liver transplantation. Liver Transpl 2006; 12:
104961
11 Pugh RNH, Murray-Lyon IM, Dawson JL, Pietroni MC, Williams R.
Transection of the oesophagus for bleeding oesophageal
varices. Br J Surg 1973; 60: 6468

i57

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

astrocytes that detoxify it to glutamine. An increased concentration of intracellular glutamine leads to swollen astrocytes
with reduced ability to regulate neurotransmission.80 101 152
155
High serum ammonia levels are present in patients with
hepatic encephalopathy, even though the degree of elevation
of ammonia does not correlate with neurological dysfunction.
Other factors, such as hyponatraemia, gastrointestinal bleeding, and infection, may contribute to the development of
hepatic encephalopathy.101 151 156 Left untreated, cerebral
oedema can progress to intracranial hypertension and brain
herniation. Multiple grading scales assess the severity of
hepatic encephalopathy. The most common scales are the
West Haven Criteria (Conn Score) which has grades 0 through
4 and the Glasgow coma score.157 158
Initial therapy for hepatic encephalopathy identifies
and treats reversible triggers (i.e. gastrointestinal bleeding,
hypokalaemia, alkalaemia, hypoglycaemia, hypovolaemia,
and infection) of this neuropsychiatric syndrome and considers
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
accepted that ammonia is a factor.159 Non-absorbable disaccharides such as lactulose acidify the gut environment to decrease absorption of ammonia from the gut via catharsis.152
Excessive dosing of lactulose causes dehydration however.
Oral antibiotics (rifaximin, neomycin, vancomycin, paramomycin, or metronidazole) reduce ammonia-producing enteric bacteria.156 Rifaximin, in combination with lactulose, might reduce
episodes of hepatic encephalopathy.151 156
Patients who develop fulminant hepatic failure are at risk for
hepatic encephalopathy, cerebral oedema with increased
intracranial pressure (ICP), and herniation. In cases of altered
mental status, a head CT scan is often indicated to evaluate
intracranial bleeding, herniation, the extent of cerebral
oedema, or both. ICP monitoring should be seriously considered for patients with fulminant hepatic failure and encephalopathy, even given the bleeding risks associated with invasive
monitors. Bleeding risk seems to be the greatest deterrent to
ICP monitoring.
ICP elevations to .25 mm Hg should be treated with mannitol to achieve hyperosmolarity. Preoperative hyperventilation
has not been shown to improve outcome and corticosteroids
are not indicated. Pentobarbital coma can be used for patients
unresponsive to mannitol, but can worsen cerebral hypoperfusion by causing systemic hypotension. Many centres consider sustained cerebral hypoperfusion (cerebral perfusion
pressure ,40 mm Hg) a contraindication to transplant because
of the high risk for brain death. In the future, transcranial
Doppler ultrasonography, near-infrared spectrophotometry,
and measurements of extracellular lactate via cerebral microdialysis could offer means of monitoring for elevated ICP in
these patients.160 161 For anaesthesia, drugs or conditions that
exacerbate elevations in ICP should be avoided. Recent evidence
shows that moderate hypothermia (32338C) can reduce cerebral oedema and intracerebral hypertension; however, the sideeffects of these manoeuvres are greater risk of infection, cardiac
dysrhythmias, and coagulopathy.162 166

BJA

i58

33 Tachibana M, Kotoh T, Kinugasa S, et al. Esophageal cancer with


cirrhosis of the liver: results of esophagectomy in 18 consecutive
patients. Ann Surg Oncol 2000; 7: 758 63
34 RN S. Anesthetic concerns for the patient with liver disease. Anesth
Analg 2008; 106: 1 7
35 Prescott LF, Forrest JAH, Adjepon-Yamoah KK, Finlayson NDC. Drug
metabolism in liver disease. J Clin Pathol 1975; 28: 62 5
36 Howden CW, Birnie GG, Brodie MJ. Drug metabolism in liver
disease. Pharmacol Ther 1989; 40: 43974
37 Hoyumpa AM, Schenker S. Is glucuronidation truly preserved in
patients with liver disease? Hepatology 1991; 13: 786 95
38 Delco F, Tchambaz L, Schlienger R, Drewe J, Krahenbuhl S. Dose adjustment in patients with liver disease. Drug Saf 2005; 28: 529 45
39 MacGilchrist AJ, Birnie GG, Cook A, et al. Pharmacokinetics and
pharmacodynamics of intravenous midazolam in patients with
severe alcoholic cirrhosis. Gut 1986; 27: 1905
40 Ochs HR, Greenblatt DJ, Eckardt B, Harmatz JS, Shader RI.
Repeated diazepam dosing in cirrhotic patients: cumulation and
sedation. Clin Pharmacol Ther 1983; 33: 4716
41 Kraus JW, Desmond PV, Marshall JP, Johnson RF, Schenker S,
Wilkinson GR. Effects of aging and liver disease on disposition of
lorazepam. Clin Pharmacol Ther 1978; 24: 411 9
42 Bansky G, Meier PJ, Riederer E, Walser H, Ziegler WH, Schmid M.
Effects of the benzodiazepine receptor antagonist flumazenil in
hepatic encephalopathy in humans. Gastroenterology 1989; 97:
744 50
43 Servin F, Cockshott ID, Farinotti R, Haberer JP, Winckler C,
Desmonts JM. Pharmacokinetics of propofol infusions in patients
with cirrhosis. Br J Anaesth 1990; 65: 177 83
44 Tegeder I, Lotsch J, Geisslinger G. Pharmacokinetics of opioids in
liver disease. Clin Pharmacokinet 1999; 37: 1740
45 Laidlaw J, Read AE, Sherlock S. Morphine tolerance in hepatic cirrhosis. Gastroenterology 1961; 40: 389 96
46 Hasselstrom J, Eriksson S, Persson A, Rane A, Svensson JO,
Sawe J. The metabolism and bioavailability of morphine in
patients with severe liver cirrhosis. Br J Clin Pharmacol 1990; 29:
289 97
47 Mazoit JX, Sandouk P, Zetlaoui P, Scherrmann JM. Pharmacokinetics of unchanged morphine in normal and cirrhotic subjects.
Anesth Analg 1987; 66: 2938
48 Devlin JW, Roberts RJ. Pharmacology of commonly used analgesics and sedatives in the ICU: benzodiazepines, propofol, and
opioids. Crit Care Clin 2009; 25: 431 49, vii
49 Hunter JM, Parker CJ, Bell CF, Jones RS, Utting JE. The use of different doses of vecuronium in patients with liver dysfunction. Br J
Anaesth 1985; 57: 758 64
50 Khalil M, DHonneur G, Duvaldestin P, Slavov V, De Hys C, Gomeni R.
Pharmacokinetics and pharmacodynamics of rocuronium in
patients with cirrhosis. Anesthesiology 1994; 80: 1241 7
51 Lebrault C, Berger JL, DHollander AA, Gomeni R, Henzel D,
Duvaldestin P. Pharmacokinetics and pharmacodynamics of
vecuronium (ORG NC 45) in patients with cirrhosis. Anesthesiology
1985; 62: 601 5
52 Magorian T, Wood P, Caldwell J, et al. The pharmacokinetics and
neuromuscular effects of rocuronium bromide in patients with
liver disease. Anesth Analg 1995; 80: 7549
53 Servin FS, Lavaut E, Kleef U, Desmonts JM. Repeated doses of
rocuronium bromide administered to cirrhotic and control
patients receiving isoflurane. A clinical and pharmacokinetic
study. Anesthesiology 1996; 84: 1092100
54 van Miert MM, Eastwood NB, Boyd AH, Parker CJ, Hunter JM. The
pharmacokinetics and pharmacodynamics of rocuronium in

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

12 Garrison RN, Cryer HM, Howard DA, Polk HC Jr. Clarification of risk
factors for abdominal operations in patients with hepatic cirrhosis.
Ann Surg 1984; 199: 64855
13 Mansour A, Watson W, Shayani V, Pickleman J. Abdominal operations in patients with cirrhosis: still a major surgical challenge.
Surgery 1997; 122: 730 5; discussion 56
14 Telem DA, Schiano T, Goldstone R, et al. Factors that predict
outcome of abdominal operations in patients with advanced cirrhosis. Clin Gastroenterol Hepatol 2010; 8: 451 7, quiz e58
15 Kamath PS, Wiesner RH, Malinchoc M, et al. A model to predict survival in patients with end-stage liver disease. Hepatology 2001; 33:
464 70
16 Said A, Williams J, Holden J, et al. Model for end stage liver disease
score predicts mortality across a broad spectrum of liver disease.
J Hepatol 2004; 40: 897903
17 Befeler AS, Palmer DE, Hoffman M, Longo W, Solomon H, Di
Bisceglie AM. The safety of intra-abdominal surgery in patients
with cirrhosis: model for end-stage liver disease score is superior
to Child Turcotte Pugh classification in predicting outcome.
Arch Surg 2005; 140: 6504; discussion 5
18 Northup PG, Wanamaker RC, Lee VD, Adams RB, Berg CL. Model for
end-stage liver disease (MELD) predicts nontransplant surgical
mortality in patients with cirrhosis. Ann Surg 2005; 242: 244 51
19 Perkins L, Jeffries M, Patel T. Utility of preoperative scores for predicting morbidity after cholecystectomy in patients with cirrhosis.
Clin Gastroenterol Hepatol 2004; 2: 11238
20 Hanje AJ, Patel T. Preoperative evaluation of patients with liver
disease. Nat Clin Pract Gastroenterol Hepatol 2007; 4: 26676
21 Kim WR, Biggins SW, Kremers WK, et al. Hyponatremia and mortality among patients on the liver-transplant waiting list. N Engl J Med
2008; 359: 101826
22 Gelman SI. Disturbances in hepatic blood flow during anesthesia
and surgery. Arch Surg 1976; 111: 8813
23 Torrance HB. Liver blood flow during operations on the upper
abdomen. J Roy Coll Surg Edinb 1957; 2: 216 28
24 Neeff H, Mariaskin D, Spangenberg HC, Hopt UT, Makowiec F.
Perioperative mortality after non-hepatic general surgery in
patients with liver cirrhosis: an analysis of 138 operations in the
2000 s using Child and MELD scores. J Gastrointest Surg 2011;
15: 1 11
25 Aranha GV, Sontag SJ, Greenlee HB. Cholecystectomy in cirrhotic
patients: a formidable operation. Am J Surg 1982; 143: 55 60
26 Belghiti J, Cherqui D, Langonnet F, Fekete F. Esophagogastrectomy
for carcinoma in cirrhotic patients. Hepato-Gastroenterology
1990; 37: 388 91
27 Bloch RS, Allaben RD, Walt AJ. Cholecystectomy in patients with
cirrhosis. A surgical challenge. Arch Surg 1985; 120: 669 72
28 Demetriades D, Constantinou C, Salim A, Velmahos G, Rhee P,
Chan L. Liver cirrhosis in patients undergoing laparotomy for
trauma: effect on outcomes. J Am Coll Surg 2004; 199: 538 42
29 Douard R, Lentschener C, Ozier Y, Dousset B. Operative risks of digestive surgery in cirrhotic patients. Gastroenterol Clin Biol 2009;
33: 55564
30 Filsoufi F, Salzberg SP, Rahmanian PB, et al. Early and late outcome
of cardiac surgery in patients with liver cirrhosis. Liver Transpl
2007; 13: 990 5
31 Klemperer JD, Ko W, Krieger KH, et al. Cardiac operations in
patients with cirrhosis. Ann Thorac Surg 1998; 65: 85 7
32 Suman A, Barnes DS, Zein NN, Levinthal GN, Connor JT, Carey WD.
Predicting outcome after cardiac surgery in patients with cirrhosis:
a comparison of Child Pugh and MELD scores. Clin Gastroenterol
Hepatol 2004; 2: 719 23

Kiamanesh et al.

BJA

Monitoring and managing hepatic disease in anaesthesia

55

56

57

58

59

60

62
63

64
65
66

67
68

69

70
71
72
73
74

75

76

77

78

79

80

81

82

83

84
85
86
87

88

89
90

91
92

93

94

95

96

shunt (TIPS) in the management of portal hypertension: update


2009. Hepatology 2010; 51: 306
DeGasperi A, Corti A, Corso R, et al. Transjugular intrahepatic portosystemic shunt (TIPS): the anesthesiological point of view
after 150 procedures managed under total intravenous anesthesia. J Clin Monit Comput 2009; 23: 341 6
Gaba RC, Khiatani VL, Knuttinen MG, et al. Comprehensive review
of TIPS technical complications and how to avoid them. Am J
Roentgenol 2011; 196: 67585
Colombato LA, Spahr L, Martinet JP, et al. Haemodynamic adaptation two months after transjugular intrahepatic portosystemic
shunt (TIPS) in cirrhotic patients. Gut 1996; 39: 6004
Merli M, Valeriano V, Funaro S, et al. Modifications of cardiac function in cirrhotic patients treated with transjugular intrahepatic
portosystemic shunt (TIPS). Am J Gastroenterol 2002; 97: 142 8
Iwakiri Y, Groszmann RJ. The hyperdynamic circulation of chronic
liver diseases: from the patient to the molecule. Hepatology 2006;
43: S121 31
Hennenberg M, Trebicka J, Sauerbruch T, Heller J. Mechanisms of
extrahepatic vasodilation in portal hypertension. Gut 2008; 57:
130014
Vaughan RB, Angus PW, Chin-Dusting JPF. Evidence for altered
vascular responses to exogenous endothelin-1 in patients with
advanced cirrhosis with restoration of the norrmal vasoconstrictor
response following successful liver transplantation. Gut 2003; 52:
150510
Hall TH, Dhir A. Anesthesia for liver transplantation. Semin Cardiothorac Vasc Anesth 2013; 17: 18094
Moller S, Hove JD, Dixen U, Bendtsen F. New insights into cirrhotic
cardiomyopathy. Int J Cardiol 2013; 167: 11018
Zardi EM, Abbate A, Zardi DM, et al. Cirrhotic cardiomyopathy. J Am
Coll Cardiol 2010; 56: 539 49
Biancofiore G, Mandell MS, Rocca GD. Perioperative considerations
in patients with cirrhotic cardiomyopathy. Curr Opin Anaesthesiol
2010; 23: 128 32
Giusca S, Jinga M, Jurcut C, Jurcut R, Serban M, Ginghina C. Portopulmonary hypertension: from diagnosis to treatment. Eur J
Intern Med 2011; 22: 441 7
Stauber RE, Olschewski H. Portopulmonary hypertension: short
review. Eur J Gastroenterol Hepatol 2010; 22: 385 90
Porres-Aguilar M, Altamirano JT, Torre-Delgadillo A, Charlton MR,
Duarte-Rojo A. Portopulmonary hypertension and hepatopulmonary syndrome: a clinician-oriented overview. Eur Respir Rev
2012; 21: 223 33
Kawut SM, Krowka MJ, Trotter JF, et al. Clinical risk factors for portopulmonary hypertension. Hepatology 2008; 48: 196203
Chen HS, Xing SR, Xu WG, et al. Portopulmonary hypertension in cirrhotic patients: prevalence, clinical features and risk factors. Exp
Ther Med 2013; 5: 81924
Krowka MJ, Plevak DJ, Findlay JY, Rosen CB, Wiesner RH, Krom RA.
Pulmonary hemodynamics and perioperative cardiopulmonaryrelated mortality in patients with portopulmonary hypertension
undergoing liver transplantation. Liver Transpl 2000; 6: 443 50
Murray KF, Carithers RL Jr, AASLD. AASLD practice guidelines:
evaluation of the patient for liver transplantation. Hepatology
2005; 41: 1407 32
Ramsay MA, Simpson BR, Nguyen AT, Ramsay KJ, East C,
Klintmalm GB. Severe pulmonary hypertension in liver transplant
candidates. Liver Transpl 1997; 3: 494 500
Marik PE, Baram M, Vahid B. Does central venous pressure predict
fluid responsiveness? A systematic review of the literature and the
tale of seven mares. Chest 2008; 134: 1728

i59

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

61

patients with hepatic cirrhosis. Br J Clin Pharmacol 1997; 44:


139 44
Viby-Mogensen J, Hanel HK. Prolonged apnoea after suxamethonium: an analysis of the first 225 cases reported to the Danish
Cholinesterase Research Unit. Acta Anaesthesiol Scand 1978; 22:
371 80
Gatecel C, Losser MR, Payen D. The postoperative effects of halothane versus isoflurane on hepatic artery and portal vein blood
flow in humans. Anesth Analg 2003; 96: 7405, table of contents
ORiordan J, OBeirne HA, Young Y, Bellamy MC. Effects of desflurane and isoflurane on splanchnic microcirculation during major
surgery. Br J Anaesth 1997; 78: 95 6
Borromeo CJ, Stix MS, Lally A, Pomfret EA. Epidural catheter and
increased prothrombin time after right lobe hepatectomy for
living donor transplantation. Anesth Analg 2000; 91: 1139 41
Schumann R, Zabala L, Angelis M, Bonney I, Tighiouart H, Carr DB.
Altered hematologic profiles following donor right hepatectomy
and implications for perioperative analgesic management. Liver
Transpl 2004; 10: 3638
Siniscalchi A, Begliomini B, De Pietri L, et al. Increased prothrombin
time and platelet counts in living donor right hepatectomy: implications for epidural anesthesia. Liver Transpl 2004; 10: 1144 9
Tsui SL, Yong BH, Ng KF, Yuen TS, Li CC, Chui KY. Delayed epidural
catheter removal: the impact of postoperative coagulopathy.
Anaesth Intensive Care 2004; 32: 6306
Hulshoff A, Schricker T, Elgendy H, Hatzakorzian R, Lattermann R.
Albumin synthesis in surgical patients. Nutrition 2013; 29: 703 7
Vincent JL, Dubois MJ, Navickis RJ, Wilkes MM. Hypoalbuminemia
in acute illness: is there a rationale for intervention? A
meta-analysis of cohort studies and controlled trials. Ann Surg
2003; 237: 319 34
Giannini EG, Testa R, Savarino V. Liver enzyme alteration: a guide
for clinicians. Can Med Assoc J 2005; 172: 367 79
Limdi JK, Hyde GM. Evaluation of abnormal liver function tests.
Postgrad Med J 2003; 79: 307 12
Hoekstra LT, de Graaf W, Nibourg GA, et al. Physiological and biochemical basis of clinical liver function tests: a review. Ann Surg
2013; 257: 27 36
Rochling FA. Evaluation of abnormal liver tests. Clin Cornerstone
2001; 3: 112
Dib N, Oberti F, Cales P. Current management of the complications
of portal hypertension: variceal bleeding and ascites. Can Med
Assoc J 2006; 174: 143343
Berzigotti A, Seijo S, Reverter E, Bosch J. Assessing portal hypertension in liver disease. Expert Rev Gastroenterol Hepatol 2013; 7:
141 55
Shibayama Y, Nakata K. Localization of increased hepatic vascular
resistance in liver cirrhosis. Hepatology 1985; 5: 6438
Groszmann RJ, Abraldes JG. Portal hypertensionfrom bench to
bedside. J Clin Gastroenterol 2005; 39: S12530
Wiklund RA. Preoperative preparation of patients with advanced
liver disease. Crit Care Med 2004; 32: S106 15
Garcia-Tsao G, Bosche J. Management of varices and variceal
hemorrhage in cirrhosis. N Engl J Med 2010; 362: 82332
Villanueva C, Colomo A, Bosch A, et al. Transfusion strategies for
acute upper gastrointestinal bleeding. N Engl J Med 2013; 368:
11 21
Gines P, Tito L, Arroyo V, et al. Randomized comparative study of
therapeutic paracentesis with and without intravenous albumin
in cirrhosis. Gastroenterology 1988; 94: 1493502
Boyer TD, Haskal ZJ, American Association for the Study of Liver
Diseases. The role of transjugular intrahepatic portosystemic

BJA

i60

119 Quittnat F, Gross P. Vaptans and the treatment of water-retaining


disorders. Semin Nephrol 2006; 26: 234 43
120 Ayus JC, Krothapalli RK, Arieff AI. Treatment of symptomatic hyponatremia and its relation to brain damage. A prospective study.
N Engl J Med 1987; 317: 1190 5
121 Hantman D, Rossier B, Zohlman R, Schrier R. Rapid correction of
hyponatremia in the syndrome of inappropriate secretion of antidiuretic hormone. An alternative treatment to hypertonic saline.
Ann Intern Med 1973; 78: 8705
122 Sterns RH, Riggs JE, Schochet SS Jr. Osmotic demyelination syndrome following correction of hyponatremia. N Engl J Med 1986;
314: 153542
123 Nordlie RC, Foster JD, Lange AJ. Regulation of glucose production
by the liver. Annu Rev Nutr 1999; 19: 379 406
124 Del Vecchio Blanco C, Gentile S, Marmo R, Carbone L, Coltori M.
Alterations of glucose metabolism in chronic liver disease.
Diabetes Res Clin Pract 1990; 8: 29 36
125 Takikawa Y, Harada M, Wang T, Suzuki K. Usefulness and accuracy of the international normalized ratio and activity percent
of prothrombin time in patients with liver disease. Hepatol Res
2013 Advance Access published on 21 February 2013. doi:
10.1111/hepr.12093
126 Tripodi A, Mannucci PM. The coagulopathy of chronic liver disease.
N Engl J Med 2011; 365: 147 56
127 Northup PG, Caldwell SH. Coagulation in liver disease: a guide for
the clinician. Clin Gastroenterol 2013; 11: 106474
128 Tripodi A, Caldwell SH, Hoffman M, Trotter JF, Sanyal AJ. Review
article: the prothrombin time test as a measure of bleeding risk
and prognosis in liver disease. Aliment Pharmacol Ther 2007; 26:
141 8
129 Dalmau A, Sabate A, Aparicio I. Hemostasis and coagulation monitoring and management during liver transplantation. Curr Opin
Organ Transplant 2009; 14: 28690
130 Krzanicki D, Sugavanam A, Mallett S. Intra-operative hypercoagulability during liver transplantation as demonstrated by thromboelastography. Liver Transpl 2013; 19: 85261
131 Findlay JY, Rettke SR. Poor prediction of blood transfusion requirements in adult liver transplantations from preoperative variables.
J Clin Anesth 2000; 12: 31923
132 Massicotte L, Beaulieu D, Thibeault L, et al. Coagulation defects do
not predict blood product requirements during liver transplantation. Transplantation 2008; 85: 956 62
133 Steib A, Freys G, Lehmann C, Meyer C, Mahoudeau G. Intraoperative blood losses and transfusion requirements during adult liver
transplantation remain difficult to predict. Can J Anaesth 2001;
48: 10759
134 Lisman T, Caldwell SH, Burroughs AK, et al. Hemostasis and thrombosis in patients with liver disease: the ups and downs. J Hepatol
2010; 53: 362 71
135 Martinez J, MacDonald KA, Palascak JE. The role of sialic acid in
the dysfibrinogenemia associated with liver disease: distribution of
sialic acid on the constituent chains. Blood 1983; 61: 1196202
136 Caldwell SH, Hoffman M, Lisman T, et al. Coagulation disorders
and hemostasis in liver disease: pathophysiology and critical assessment of current management. Hepatology 2006; 44: 103946
137 Colucci M, Binetti BM, Branca MG, et al. Deficiency of thrombin activatable fibrinolysis inhibitor in cirrhosis is associated with
increased plasma fibrinolysis. Hepatology 2003; 38: 2307
138 Lisman T, Leebeek FW, Mosnier LO, et al. Thrombin-activatable
fibrinolysis inhibitor deficiency in cirrhosis is not associated
with increased plasma fibrinolysis. Gastroenterology 2001; 121:
131 9

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

97 Burger-Klepp U, Karatosic R, Thum M, et al. Transesophageal echocardiography during orthotopic liver transplantation in patients
with esophagoastric varices. Transplantation 2012; 94: 192 6
98 Suriani RJ, Cutrone A, Feierman D, Konstadt S. Intraoperative
transesophageal echocardiography during liver transplantation.
J Cardiothorac Vasc Anesth 1996; 10: 699707
99 Hemprich U, Papadakos PJ, Lachmann B. Respiratory failure and
hypoxemia in the cirrhotic patient including hepatopulmonary
syndrome. Curr Opin Anaesthesiol 2010; 23: 1338
100 Fallon MB, Abrams GA. Pulmonary dysfunction in chronic liver
disease. Hepatology 2000; 32: 859 65
101 Bernal W, Auzinger G, Dhawan A, Wendon J. Acute liver failure.
Lancet 2010; 376: 190210
102 Rodriguez-Roisin R, Krowka MJ, Herve P, Fallon MB, Committee
ERSTFP-HVDS. Pulmonary-hepatic vascular disorders (PHD). Eur
Respir J 2004; 24: 86180
103 Rodriguez-Roisin R, Krowka MJ. Hepatopulmonary syndromea
liver-induced lung vascular disorder. N Engl J Med 2008; 358:
2378 87
104 Zhang J, Fallon MB. Hepatopulmonary syndrome: update on
pathogenesis and clinical features. Nat Rev Gastroenterol
Hepatol 2012; 9: 539 49
105 Tanikella R, Fallon MB. Hepatopulmonary syndrome and liver
transplantation: who, when and where? Hepatology 2013; 57:
2097 9
106 Krowka MJ, Mandell MS, Ramsay MA, et al. Hepatopulmonary syndrome and portopulmonary hypertension: a report of the multicenter liver transplant database. Liver Transpl 2004; 10: 174 82
107 Hartleb M, Gutkowski K. Kidneys in chronic liver diseases. World J
Gastroenterol 2012; 18: 3035 49
108 Meltzer J, Brentjens TE. Renal failure in patients with cirrhosis:
hepatorenal syndrome and renal support strategies. Curr Opin
Anaesthesiol 2010; 23: 13944
109 Gines P, Schrier RW. Renal failure in cirrhosis. N Engl J Med 2009;
361: 127990
110 Steadman RH, Van Rensburg A, Kramer DJ. Transplantation for
acute liver failure: perioperative management. Curr Opin Organ
Transplant 2010; 15: 36873
111 Anderson CL, Saad WE, Kalagher SD, et al. Effect of transjugular
intrahepatic portosystemic shunt placement on renal function:
a 7-year, single-center experience. J Vasc Interv Radiol 2010; 21:
1370 6
112 Gine`s P, Guevara M, Arroyo V, Rodes J. Hepatorenal syndrome.
Lancet 2003; 362: 181927
113 Salerno F, Gerbes A, Gines P, Wong F, Arroyo V. Diagnosis, prevention and treatment of hepatorenal syndrome in cirrhosis. Postgrad
Med J 2008; 84: 66270
114 Gluud LL, Christensen K, Christensen E, Krag A. Terlipressin for
hepatorenal syndrome. Cochrane Database Syst Rev 2012; 9:
CD005162
115 Arroyo V, Terra C, Gines P. New treatments of hepatorenal syndrome. Semin Liver Dis 2006; 26: 254 64
116 Gonwa TA, Morris CA, Goldstein RM, Husberg BS, Klintmalm GB.
Long-term survival and renal function following liver transplantation in patients with and without hepatorenal syndromeexperience in 300 patients. Transplantation 1991; 51: 42830
117 Seu P, Wilkinson AH, Shaked A, Busuttil RW. The hepatorenal
syndrome in liver transplant recipients. Am Surg 1991; 57:
806 9
118 Gines P, Guevara M. Hyponatremia in cirrhosis: pathogenesis,
clinical significance, and management. Hepatology 2008; 48:
1002 10

Kiamanesh et al.

BJA

Monitoring and managing hepatic disease in anaesthesia

153

154
155

156
157

158
159

160

161

162

163

164

165

166

in fulminant hepatic failure. Curr Opin Anaesthesiol 2010; 23:


1217
Bajaj JS, Pinkerton SD, Sanyal AJ, Heuman DM. Diagnosis and
treatment of minimal hepatic encephalopathy to prevent motor
vehicle accidents: a cost-effectiveness analysis. Hepatology
2012; 55: 1164 71
Riordan SM, Williams R. Treatment of hepatic encephalopathy.
N Engl J Med 1997; 337: 473 9
Bjerring PN, Eefsen M, Hansen BA, Larsen FS. The brain in acute liver
failure. A tortuous path from hyperammonemia to cerebral
edema. Metab Brain Dis 2009; 24: 5 14
Bass N, Mullen K, Sanyal A, et al. Rifaximin treatment in hepatic encephalopathy. N Engl J Med 2010; 362: 1071 81
Sakamoto M, Perry W, Hilsabeck RC, Barakat F, Hassanein T.
Assessment and usefulness of clinical scales for semiquantification
of overt hepatic encephalopathy. Clin Liver Dis 2012; 16: 2742
Blei AT, Cordoba J. Hepatic encephalopathy. Am J Gastroenterol
2001; 96: 1968 76
Ong JP, Aggarwal A, Krieger D, et al. Correlation between ammonia
levels and the severity of hepatic encephalopathy. Am J Med 2003;
114: 18893
Nielsen HB, Tofteng F, Wang LP, Larsen FS. Cerebral oxygenation
determined by near-infrared spectrophotometry in patients
with fulminant hepatic failure. J Hepatol 2003; 38: 188 92
Tofteng F, Jorgensen L, Hansen BA, Ott P, Kondrup J, Larsen FS.
Cerebral microdialysis in patients with fulminant hepatic failure.
Hepatology 2002; 36: 1333 40
Jalan R, O Damink SW, Deutz NE, Lee A, Hayes PC. Moderate hypothermia for uncontrolled intracranial hypertension in acute liver
failure. Lancet 1999; 354: 1164 8
Jalan R, Olde Damink SW, Deutz NE, et al. Moderate hypothermia
prevents cerebral hyperemia and increase in intracranial pressure
in patients undergoing liver transplantation for acute liver failure.
Transplantation 2003; 75: 2034 9
Jalan R, Olde Damink SW, Deutz NE, Hayes PC, Lee A. Moderate
hypothermia in patients with acute liver failure and
uncontrolled intracranial hypertension. Gastroenterology 2004;
127: 133846
Dmello D, Cruz-Flores S, Matuschak GM. Moderate hypothermia
with intracranial pressure monitoring as a therapeutic paradigm
for the management of acute liver failure: a systematic review. Intensive Care Med 2010; 36: 2103
Stravitz RT, Larsen FS. Therapeutic hypothermia for acute liver
failure. Crit Care Med 2009; 37: S25864

Handling editor: H. C. Hemmings

i61

Downloaded from http://bja.oxfordjournals.org/ by guest on July 7, 2016

139 Lisman T, Leebeek FW. Hemostatic alterations in liver disease: a


review on pathophysiology, clinical consequences, and treatment. Dig Surg 2007; 24: 2508
140 Mallett SV, Chowdary P, Burroughs AK. Clinical utility of viscoelastic
tests of coagulation in patients with liver disease. Liver Int 2013;
33: 961 74
141 Tripodi A, Primignani M, Chantarangkul V, et al. The coagulopathy
of cirrhosis assessed by thromboelastometry and its correlation
with conventional coagulation parameters. Thromb Res 2009;
124: 1326
142 Pham HP, Shaz BH. Update on massive transfusion. Br J Anaesth
2013; 111 (Suppl. 1): i71 i82
143 Sihler KC, Napolitano LM. Complications of massive transfusion.
Chest 2010; 137: 20920
144 Friederich PW, Henny CP, Messelink EJ, et al. Effect of recombinant
activated factor VII on perioperative blood loss in patients undergoing retropubic prostatectomy: a double-blind placebocontrolled randomised trial. Lancet 2003; 361: 2015
145 Lodge JP, Jonas S, Jones RM, et al. Efficacy and safety of repeated
perioperative doses of recombinant factor VIIa in liver transplantation. Liver Transpl 2005; 11: 973 9
146 Bernstein DE, Jeffers L, Erhardtsen E, et al. Recombinant factor
VIIa corrects prothrombin time in cirrhotic patients: a preliminary
study. Gastroenterology 1997; 113: 19307
147 Jeffers L, Chalasani N, Balart L, Pyrsopoulos N, Erhardtsen E. Safety
and efficacy of recombinant factor VIIa in patients with liver
disease undergoing laparoscopic liver biopsy. Gastroenterology
2002; 123: 118 26
148 Levy JH, Fingerhut A, Brott T, Langbakke IH, Erhardtsen E, Porte RJ.
Recombinant factor VIIa in patients with coagulopathysecondary
to anticoagulant therapy, cirrhosis, or severe traumatic injury:
review of safety profile. Transfusion 2006; 46: 91933
149 Shao YF, Yang JM, Chau GY, et al. Safety and hemostatic effect of
recombinant activated factor VII in cirrhotic patients undergoing
partial hepatectomy: a multicenter, randomized, double-blind,
placebo-controlled trial. Am J Surg 2006; 191: 245 9
150 Cohen B, Matot I. Aged Erythrocytesa fine wine or sour grapes?.
Br J Anaesth 2013; 111 (Suppl. 1): i62 i70
151 Teperman LW, Peyregne VP. Considerations on the impact of
hepatic encephalopathy treatments in the pretransplant
setting. Transplantation 2010; 89: 7718
152 Zafirova Z, OConnor M. Hepatic encephalopathy: current management strategies and treatment, including management
and monitoring of cerebral edema and intracranial hypertension

Potrebbero piacerti anche