Вы находитесь на странице: 1из 14

Epidemiology, Pathophysiology, and Management

of Hepatorenal Syndrome

D1X XAhmed Adel Amin, MD, D2X X*,† D3X XEman Ibrahim Alabsawy, MD, *D4X X ,‡ Rajiv
D5X X Jalan, PhD, D6X X* and
D7X XAndrew Davenport, MDD§8X X

Summary: Acute kidney injury (AKI) is a common presentation in patients with advanced cirrhosis hospitalized with
acute decompensation. A new revised classification now divides AKI in cirrhotic patients into two broad subgroups:
hepatorenal syndrome AKI (HRS AKI) and non−hepatorenal syndrome AKI (non-HRS AKI). HRS AKI represents
the end-stage complication of decompensated cirrhosis with severe portal hypertension and is characterized by
worsening of renal function in the absence of prerenal azotemia, nephrotoxicity, and intrinsic renal disease. Non-
HRS AKI may be caused by prerenal hypoperfusion, bile acid nephropathy, nephrotoxicity, or acute parenchymal
insult. There have been several mechanisms proposed to explain the pathophysiology of HRS AKI and non-HRS
AKI, and a number of biomarkers have been suggested to aid in differentiation between these types of AKI and to
act as prognostic indicators. The standard of care clinical management for patients with HRS AKI is to exclude other
etiologies of AKI, followed by volume expansion with human albumin solution and then the introduction of vasopres-
sors. However, some 40% of patients treated for HRS AKI fail to respond. In this review, we discuss the current and
recent data about classification, pathophysiology, and management of AKI in general, with specific insight about the
treatment of HRS AKI.
Semin Nephrol 39:17−30 Ó 2018 Elsevier Inc. All rights reserved.
Keywords: Cirrhosis, hepatorenal syndrome, acute kidney injury, acute-on-chronic liver failure (ACLF)

R
enal dysfunction is a common finding in patients leads to marked splanchnic vasodilation. This causes
with advanced liver disease with cirrhosis and activation of both the renin-angiotensin-aldosterone sys-
portal hypertension admitted to the hospital, and tem and the sympathetic nervous system, leading to
is associated with increased morbidity and mortality. intense renal vasoconstriction, which then plays a major
Typically, the cause of renal dysfunction is multifactorial role in the pathogenesis of HRS AKI.3,4 In addition,
and, as such, specific management strategies are limited advanced portal hypertension leads to greater shear
because the exact mechanisms have not been fully eluci- stress in the splanchnic vessels, increasing bacterial
dated. Within the spectrum of acute renal dysfunction in translocation from the bowel,4 which is associated with
patients with cirrhosis, hepatorenal syndrome (HRS) car- increased generation of nitric oxide (NO) and prostacy-
ries the most ominous prognosis because it usually clins. These potent splanchnic vasodilators cause pool-
denotes a background of severe portal hypertension and ing of blood and decrease the effective circulating
circulatory dysfunction. HRS acute kidney injury (AKI) systemic blood volume.5 Initially, these effects are com-
accounts for approximately 11% of AKI in hospitalized pensated by hyperdynamic circulation with increased
cirrhotic patients with refractory ascites, and is associ- cardiac output, resulting from an increase in both stroke
ated with high mortality.1,2 volume and heart rate, but as the severity of the under-
In advanced cirrhosis, portal hypertension results in a lying liver disease progresses, then cardiac output does
profound hemodynamic derangement, which in turn not increase sufficiently to compensate for the degree of
vasodilation.6,7
*University College London Institute for Liver and Digestive Health, Historically, most patients admitted with renal dys-
Division of Medicine, University College London Medical School, function in the context of chronic liver disease and asci-
Royal Free Hospital, London, UK tes without evidence of intrinsic renal disease were
yAssiut University Hospital, Internal Medicine Department, Assiut considered to have HRS AKI until proven otherwise. In
University, Assiut, Egypt
zTropical Medicine Department, Alexandria University Hospital,
addition, the classification of HRS historically was sub-
Alexandria, Egypt divided into two types: type 1 when the deterioration in
xUniversity College London Centre for Nephrology, Division of Medi- renal function occurred over days to weeks, and type 2
cine, University College London Medical School, Royal Free Hos- when deterioration occurred over months.8 This classifi-
pital, London, UK cation now has been abandoned because it does not accu-
Financial disclosure and conflict of interest statements: none.
Address reprint requests to Andrew Davenport, MD, University Col-
rately reflect the clinical scenario. A newer classification
lege London Centre for Nephrology, Division of Medicine, Royal has been proposed by the International Club of Ascites
Free Hospital, University College London Medical School, Row- (ICA), which divides patients with cirrhosis and AKI
land Hill St, London NW3 2PF, UK. into distinct subgroups according to the underlying
E-mail: andrewdavenport@nhs.net pathology.1,9 Causes of AKI in cirrhotic patients other
0270-9295/ - see front matter
© 2018 Elsevier Inc. All rights reserved.
than HRS AKI are identified in the new classification
https://doi.org/10.1016/j.semnephrol.2018.10.002 and include prerenal causes, bile acid nephropathy, and

Seminars in Nephrology, Vol 39, No 1, January 2019, pp 17−30 17


18 A.A. Amin et al.

acute tubular injury.1 This type of AKI collectively is AKI is associated with an increased risk of mortality
referred to as non-HRS AKI. This term is different from because 2% to 31% of hospitalized cirrhotic patients
HRS AKI, which defines a functional type of AKI in the with AKI do not survive their admission.19,20 One- and
setting of normal renal parenchyma.1 Chronic kidney 12-month mortality rates reported in these patients are
disease represents a term that includes any cause that 58% and 63%, respectively.21 Even those who survive
structurally affects the renal parenchyma, including glo- their hospital admission are more prone to complica-
merulopathies and interstitial renal disease, and causes tions of cirrhosis, including ascites and hepatic
associated with comorbid diseases such as diabetes mel- encephalopathy.19,22
litus and hypertension.1,10
Patients with cirrhosis may suffer episodes of acute
hepatic decompensation, and acute-on-chronic liver fail- DEFINITION AND CLASSIFICATION
ure (ACLF) is a recently recognized clinical entity that
occurs in patients with acute hepatic decompensation. HRS now is described as a more homogeneous condition
ACLF is characterized by multiple organ failures and is with specific diagnostic features.10 Historically, HRS
associated with increased short-term mortality.11,12 AKI was classified into 2 types: type 1 HRS, which was
is an important factor that defines ACLF and it is consid- defined as rapid deterioration in kidney function over the
ered one of its major components, however, the patho- course of 2 weeks with a serum creatinine level greater
physiologic mechanisms of AKI in ACLF have not been than 2.5 mg/dL, and type 2 HRS, which was character-
fully elucidated because patients may have non-HRS ized by a progressive slower course of moderate renal
AKI or HRS AKI.10 failure and serum creatinine concentrations between 1.5
Currently, therapeutic options for HRS AKI remain and 2.5 mg/dL.23 The main problem with this older clas-
supportive and are only a bridge to liver transplantation. sification was that it relied predominantly on serum cre-
Despite the recent advances in scientific research in this atinine concentrations as the sole factor to classify HRS
field, the optimum management for patients with HRS into types 1 and 2, irrespective of the underlying pathol-
AKI remains to be established, and a better understand- ogy or etiology. Serum creatinine does not accurately
ing of the different pathophysiologic mechanisms of reflect the severity of renal impairment in cirrhotic
HRS and non-HRS AKI is required.13,14 In this review, patients because it is affected by several factors includ-
we discuss the recent updates in the pathogenesis, diag- ing assay interference with bilirubin, reduced hepatic
nosis, and management of HRS AKI in the setting of the creatine synthesis, muscle wasting, and malnutrition.24
new definitions and classification of AKI. In 2007, the Acute Kidney Injury Network proposed a
new definition of AKI,25 which then was supported by
both the Acute Dialysis Quality Initiative and the ICA in
2011.9 The Acute Kidney Injury Network’s new defini-
EPIDEMIOLOGY tion of AKI allowed the use of any increase of the abso-
AKI occurs in 25% to 50% of cirrhotic patients admitted lute values of serum creatinine from baseline by as little
to the hospital after an episode of acute decompensa- as 0.3 mg/dL (26.5 mmol/L), or any increase of serum
tion.15 AKI is either prerenal, renal parenchymal, or creatinine by 50% above the baseline within a 48-hour
obstructive in origin. Prerenal causes include hypovole- period (Table 1).9 This definition was supported by
mia and HRS AKI, and together they account for 60% to observational data linking AKI stage severity and patient
70% of all causes of AKI,16 with HRS AKI accounting survival.15,19, 26,27
for 11% to 20% of all causes of AKI.16,17 Intrinsic renal Recently, the ICA proposed a new definition and diag-
causes account for approximately 30% of all causes of nostic criteria for HRS AKI, and the older subclassifica-
AKI in cirrhotic patients, and include ischemic injury tions of type 1 and type 2 and the time limit of 2 weeks
and acute tubular necrosis, acute glomerulonephritis, and to diagnose type 1 HRS were removed.1 In addition, the
acute interstitial nephritis, whereas postrenal (obstruc- limiting threshold of a serum creatinine concentration of
tive) causes are relatively uncommon (<1%).16,18 2.5 mg/dL, which was the cornerstone for diagnosing

Table 1. International Club of Ascites Definition and Staging of AKI in Patients With Cirrhosis1,9
Definition of AKI
Increase in serum creatinine ≥0.3 mg/dL (≥26.5 mmol/L) within 48 hours; or a percentage increase in serum creatinine ≥50% from
baseline that is known, or presumed, to have occurred within the prior 7 days
Stages of AKI
Stage 1 Increase in serum creatinine ≥0.3 mg/dL (26.5 mmol/L) or an increase in serum creatinine ≥1.5-fold to 2-fold from baseline
Stage 2 Increase in serum creatinine >2-fold to 3-fold from baseline
Stage 3 Increase in serum creatinine >3-fold from baseline or ≥4.0 mg/dL (353.6 mmol/L) with an acute increase ≥0.3 mg/dL
(26.5 mmol/L) or initiation of renal replacement therapy
Epidemiology, pathophysiology, and HRS management 19

Table 2. ICA Diagnostic Criteria of HRS AKI1


compromise.29 In addition, increased vasopressin release
and local endothelin secretion contribute to reduced
Diagnosis of cirrhosis and ascites intraglomerular blood flow. Progression of liver disease
Diagnosis of AKI according to ICA AKI diagnostic criteria
No response at 48 hours of plasma volume expansion using results in further splanchnic vasodilation and renal vaso-
albumin 1 g/kg of body weight and withdrawal of diuretics constriction leading to functional renal impairment
Absence of shock (Fig. 1). Aldosterone and vasopressin cause both sodium
No current or recent use of nephrotoxic drugs
No macroscopic signs of structural kidney injury defined and water retention, which further worsen ascites.30,31
as follows: Evidence that HRS AKI is a functional disorder rather
Absence of proteinuria (<500 mg/d) than a structural disease includes the success of cadav-
Absence of microhematuria (<50 RBCs per high-power field)
Normal renal ultrasonography eric transplantation of kidneys donated from these
Abbreviation: RBC, red blood cells.
patients,32 post-mortem examination,32 and resolution of
HRS AKI after liver transplantation.33 The current clini-
cal management of HRS AKI targets splanchnic vasodi-
lation and effective volume depletion by using
HRS, was removed.1 According to this newer definition, splanchnic vasoconstrictors and albumin. However, this
HRS AKI now is defined as worsening kidney function treatment strategy—although improving renal function
in patients with advanced cirrhosis that meets the ICA- in the majority of patients34,35—still is unable to reverse
AKI criteria (Table 2); failing to respond to volume HRS AKI in approximately 40% of cases.36 This indi-
expansion with albumin; the absence of recent exposure cates that patients have either been misclassified as hav-
to nephrotoxic agents (such as aminoglycosides, nonste- ing non-HRS AKI or that other mechanisms are
roidal anti-inflammatories, or contrast media); and no involved.
evidence of shock or signs of structural kidney disease
(defined as proteinuria <500 mg/d, hematuria <50 red
blood cells per high-power field, and normal renal ultra- Cardiac Dysfunction
sonographic findings).1 Cirrhotic cardiomyopathy, a term describing the abnor-
This newer classification describes a new phenotypic malities detected in the cardiac response and function of
classification of AKI HRS in cirrhotic patients based on cirrhotic patients, can affect as many as 50% of patients.6
pathophysiologic characteristics. Non-HRS AKI now It is marked by the abnormal response to both physio-
describes other causes of AKI in cirrhotic patients other logic and pathologic stresses, with patients having a rela-
than HRS AKI, including bile salt nephropathy, prerenal tively low cardiac output for the degree of systemic
hypovolemia caused by bleeding, excessive diuretic use, vasodilation. Low cardiac output states in patients with
or any excessive fluid loss, acute tubular injury, acute cirrhosis and refractory ascites are found to be a predic-
tubular necrosis, and AKI caused by intrinsic renal tor of HRS AKI and are associated with worse prognosis.
causes such as acute interstitial nephritis.1 b-blockers prescribed to patients with ascites have been
reported to cause further deterioration in renal function
in patients with HRS AKI,37,38 most probably because of
PATHOPHYSIOLOGY OF HRS AKI their hypotensive effects.
There have been several pathophysiological explanations
proposed as to why HRS AKI occurs in patients with
Adrenal Insufficiency
advanced cirrhosis and portal hypertension.
Adrenal insufficiency is reported to affect approximately
25% of patients with decompensated cirrhosis.39 It
Splanchnic Vasodilation causes further deleterious effects on the heart by down-
The classic and traditional hypothesis for the develop- regulating b-adrenergic receptors and modulating the
ment of HRS AKI is a reduction in kidney function effects of catecholamines on myocardial contraction and
caused by severe systemic vasodilation and subsequent vascular responsiveness.40 These effects add to the
renal vasoconstriction. Increasing portal hypertension hemodynamic compromise of patients with decompen-
and shear stress on the portal blood vessels causes the sated cirrhosis.
endothelium to produce several locally acting vasodila-
tors such as NO and prostanoids.28,29 These vasodilators
act locally on the splanchnic vasculature causing intense Inflammation
vasodilation. In turn, the subsequent decrease in the Systemic inflammation is a key factor that predisposes to
effective mean arterial blood pressure leads to activation AKI in advanced cirrhotic patients, especially in associa-
of the renin-angiotensin-aldosterone axis, and visceral tion with ACLF.41 Inflammation is more likely to cause
sympathetic nervous system, to increase cardiac output non-HRS AKI rather than HRS AKI. Mortality rates are
and heart rate to compensate for this hemodynamic more than twice as high in patients with cirrhosis and
20 A.A. Amin et al.

cirrhosis

Portal hypertension

Splanchnic and Bacterial


systemic vasodilation translocation

Local and systemic


inflammation

Generation of vasodilator nitric


oxide and prostacyclins

Decreased effective arterial blood


volume (EABV)

Activation of neurohumoral
mechanisms (RAAS)

Sodium and water


Renal vasoconstriction
retention

Ascites and Decreased renal blood


hyponatremia flow

HRS

Figure 1. Pathophysiological basis of hepatorenal syndrome. Abbreviation: RAAS, renin-angiotensin-aldosterone system.

renal failure who have a systemic inflammatory response Systemic inflammation and bacterial translocation
than those without.42 Observational studies from patients have been proposed as one of the crucial mechanisms
with AKI and cirrhosis have reported that those patients leading to non-HRS AKI in patients with advanced cir-
with spontaneous bacterial peritonitis (SBP) had higher rhosis. Systemic and local intrarenal inflammation have
circulating levels of proinflammatory cytokines (includ- been described recently and suggested to be a key factor
ing interleukin 6 and tumor necrosis factor-a) compared for the development of AKI in the setting of ACLF. AKI
with those without SBP.43 in patients with ACLF is a heterogeneous disorder initi-
ated by multiple factors including infection, and is associ-
ated with varying degrees of systemic inflammation,
PATHOPHYSIOLOGY OF NON-HRS AKI which leads to multi-organ failure.44 In the Chronic Liver
Non-HRS AKI encompasses all of the other causes of Failure Acute on-Chronic Liver Failure in Cirrhosis Con-
AKI in the setting of decompensated cirrhosis other than sortium study, patients with renal failure (as defined by a
HRS AKI. This includes prerenal causes that lead to serum creatinine concentration ≥2 mg/dL) had 20% mor-
hypovolemia, such as excess diuretic use, upper gastroin- tality and mortality was significantly greater with other
testinal bleeding, and any other causes of severe fluid organ failures.11 Patients are more likely to have non-
loss, the toxic effects of bile acids on the renal tubules, HRS AKI than HRS AKI with higher organ failure scores
and other intrinsic renal causes such as acute tubular (chronic liver Failure organ failure), increasing serum bil-
injury and necrosis (ATN) and acute interstitial nephritis. irubin concentrations, and nonresolving infections
Epidemiology, pathophysiology, and HRS management 21

because these patients are more likely to fail to respond to Bile acids have been found to play a role in the patho-
supportive treatment with albumin and terlipressin.45,46 genesis of AKI through their direct toxic effects on the
In a recent study, various markers of systemic inflamma- renal tubules.54 Tubular bile acid casts have been found
tion including 29 serum cytokines and nonmercaptalbu- in renal biopsy specimens of patients with HRS AKI,54
min 2 were measured in both ACLF and non-ACLF which may be a contributing factor to the development
cirrhotic patients, and they were markedly and signifi- of AKI in patients with cirrhosis. Because bile acids
cantly increased in ACLF patients.47 Interestingly, the accumulate in patients with increased serum bilirubin
severity renal dysfunction was associated with increasing concentrations, this potentially may explain the link
markers of inflammation (interleukin 6, interleukin 8, and between higher bilirubin concentrations and worse out-
human nonmercaptalbumin 2), but not with plasma renin comes and reduced responsiveness to terlipressin therapy
or copeptin concentration.47 These findings suggest that in HRS AKI patients.46 This hypothesis is supported by
AKI in cirrhotic patients is associated with systemic animal data, which showed that the use of nor-ursodeox-
inflammation, especially in those with ACLF, rather than ycholic acid, which increases bile acid clearance in the
the more traditional hemodynamic dysfunction hypothe- gut, may help decrease renal injury in rat models of
sis of HRS AKI. cirrhosis.55
In a retrospective study, renal biopsy specimens from Even though HRS AKI and non-HRS AKI have dif-
65 cirrhotic patients with unexplained renal dysfunction, ferent pathogenic mechanisms, and are considered dif-
defined as a serum creatinine level greater than ferent subtypes of AKI, they also have overlapping
1.5 mg/dL, with no hematuria or proteinuria and with a characteristics. The fact that only 40% of patients with
normal ultrasound scan, were studied. Twenty-eight per- HRS AKI respond to treatment with terlipressin and
cent of these patients were found to have structural albumin, and that this unresponsiveness increases with
changes on their renal biopsy specimens, including time, raises the possibility that even if HRS AKI was
chronic tubulointerstitial injury, glomerular injury, and diagnosed correctly, then with persistent renal ischemia
vascular injury. These findings are of particular impor- HRS AKI then may evolve with time to non-HRS AKI,
tance because they show that patients who may appear to because ongoing renal ischemia will lead to inflamma-
have functional renal impairment also may have underly- tory changes within the renal interstitium and tubular
ing parenchymal lesions.48 Another finding was the cell death. Despite an initial response, 80% of patients
increased renal tubular expression of Toll-like receptor 4 with HRS AKI die within 3 months, and patients who
and caspase-3 in the biopsy specimens from 5 patients develop HRS AKI for more than 6 weeks typically fail
with non-HRS AKI and ACLF, whereas biopsy speci- to recover residual renal function even with liver
mens from patients with ACLF and HRS AKI did not transplantation.10
show these local inflammatory changes in the renal It should be noted that in patients with chronic comor-
tubules.48 This suggests that non-HRS AKI, unlike HRS bid conditions, such as diabetes mellitus and systemic
AKI, is associated with an acute tubular injury, as evi- hypertension, as well as patients with nonalcoholic fatty
denced by tubular cell death and increased Toll-like liver disease and chronic viral hepatitis B or C, or other
receptor 4 expression. These data have been reproduced glomerular or interstitial nephropathies, may have some
in animal experiments by administering endotoxin to degree of established parenchymal renal injury irrespec-
bile-ligated rat models of cirrhosis.49 tive of the degree of liver dysfunction.56 Patients with
Gut bacterial translocation increases systemic proin- chronic kidney disease are much more susceptible to
flammatory cytokines and lipopolysaccharides, which developing AKI when exposed to any acute renal
can directly cause renal tubular cell apoptosis through compromise.10
the caspase-mediated pathway.50 This renal tubular
injury can be attenuated markedly by the administration
of norfloxacin before endotoxin exposure in animal BIOMARKERS IN AKI
models of cirrhosis.49 In patients with cirrhosis, the use Early treatment of AKI is crucial for determining out-
of norfloxacin as primary prophylaxis for SBP was come, and therefore it is important to differentiate HRS
found not only to delay the onset of HRS AKI, but also AKI from non-HRS AKI. Biomarkers have been used
to improve 1-year survival.51 Rifaximin also has been for both identifying etiology and determining prognosis.
reported to reduce the incidence of AKI, including HRS In some cases, biomarkers can be used to determine
AKI.52 In a retrospective analysis of data from 4 which patients are suitable for specific therapies.
cohorts of patients treated for HRS AKI, the ACLF Serum creatinine concentration has been used to diag-
grade was the major determinant of response to terli- nose and assess the severity of AKI in routine clinical
pressin and albumin, with reduced response with higher practice. However, serum creatinine is determined not
grades. In addition, patient survival was lower with only by renal function, but also by dietary protein intake,
increasing ACLF grade independently of the response muscle mass and activity, and nonrenal clearance.57
to treatment.53 Moreover, most patients with advanced decompensated
22 A.A. Amin et al.

cirrhosis have ascites and fluid overload, which also reported to vary over the course of the day, and can be
affects the accurate measurement of serum creatinine affected by age, drugs, comorbidity, and other factors
concentration.58 High bilirubin levels also may affect the including smoking. There has now been standardization
accuracy of serum creatinine in plasma samples owing to of cystatin C assays, increasing the reliability of meas-
spectral effects and reacting with the assay reagents, urements. Although increased cystatin C is of prognostic
leading to a lowering of the laboratory-reported serum value, it has not yet been shown to be superior to creati-
creatinine concentration.59 Therefore, alternative enzy- nine in assessing renal function.
matic methods to measure creatinine in patients with Serum neutrophil gelatinase-associated lipocalin
high bilirubin levels are preferred to the colorimetric (NGAL) has been the most widely investigated marker
assays. However, in most countries, enzymatic creatinine among the newer biomarkers. NGAL is synthesized pre-
measurements are more expensive than the standard col- dominantly in the liver, but also is expressed by the renal
orimetric Jaffe-based picric acid methods and may not tubules after an inflammatory insult, be it ischemic, toxic,
be routinely available in all centers. or infective. Although some studies have suggested that it
A fractional excretion of urinary sodium (FENa) of differentiates HRS AKI, prerenal azotemia, and acute
less than 1% has been widely used to differentiate intrinsic kidney disease, the main limitation is the signifi-
patients with prerenal AKI, but patients with HRS AKI cant overlap of NGAL values in different types of AKI,
also may have reduced FENa. Although it has been sug- and urinary NGAL is increased with urinary tract infec-
gested that a very low FENa of <0.1% may be support- tions.65 Similarly, other urinary biomarkers, such as inter-
ive of a diagnosis of prerenal AKI in patients with leukin 18, kidney injury molecule-1, hepatic fatty acid
cirrhosis, it must be remembered that not only does the binding protein, insulin-like growth hormone 1, and tissue
ability to concentrate urinary sodium fall with age, but inhibitor of metalloproteinase 2, have not been shown to
that urinary sodium excretion also can be affected by clearly separate AKI HRS and non-HRS AKI.
diuretics and sepsis, and as such the diagnostic value of In a recent study, high levels of circulating microRNA-
FENa in routine clinical practice has limited value.60 21 and low levels of microRNA 146a and 210 were
There has been renewed interest in measuring the observed in both ATN and HRS AKI patients compared
fractional excretion of urea as a marker to differentiate with controls, with a statistically significant difference
between renal hypoperfusion and tubular injury.61 Urea between ATN and HRS. These preliminary results sug-
is filtered by the glomeruli and then reabsorbed in the gest that different microRNAs potentially could be used
proximal renal tubules and concentrated in the inner to differentiate between ATN and HRS, but such prelimi-
medulla to generate a concentration gradient,62 so any nary observational reports require further evaluation.66
renal hypoperfusion should decrease its fractional excre-
tion, whereas any tubular injury should cause an increase
in its fractional excretion. Because diuretics predomi- TREATMENT
nantly affect the ascending loop of Henle and distal Early diagnosis and rapid medical treatment is crucial
tubule, urea is less affected by the action of diuretics.60 in HRS because AKI HRS may progress to nonrevers-
In a recent cohort study performed on 50 patients with ible HRS AKI. The standard of care in the manage-
cirrhosis and ascites admitted with AKI, fractional excre- ment of HRS is based on the understanding of the
tion of urea was found to be a promising tool to differen- hemodynamic dysfunction, which underpins its patho-
tiate AKI of different causes, including HRS, prerenal genesis. Patients suffering from cirrhosis, ascites, and
azotemia, and tubular injury.61 AKI should be managed according to ICA recommen-
Recently, several different studies suggested measur- dations.67 Optimized biomarkers to differentiate HRS
ing serum cystatin C as an alternative to serum creatinine AKI from structural kidney injury and renal function
for assessing renal function and, more importantly, pre- algorithms may help to refine management.68
dicting prognosis. Cystatin C is secreted from all nucle-
ated cells in the body and freely passes through the
Volume Expansion and Removing
glomeruli, and was thought to be removed exclusively
by the kidney.63 A recent observational study that Nephrotoxic Agents
included 350 patients with cirrhosis and ascites reported Any potential nephrotoxic drugs (ie, nonsteroidal anti-
that serum cystatin C was an independent predictor of inflammatory drugs, diuretics, angiotensin-converting
mortality and HRS when compared with serum creati- enzyme inhibitors, antibiotics, and so forth) ideally
nine.64 However, cystatin C is increased in inflammatory should be minimized or stopped, and any element of
conditions, and older studies have noted a progressive hypovolemia corrected (Fig. 2).69 Intravascular volume
increase between patients with Child-Turcotte-Pugh assessment is an initial key step to ensure that hypovole-
grades from A to C, and it is unclear whether nonrenal mia is managed adequately. Accurate volume assessment
clearance of cystatin C is altered in patients with in cirrhotic patients with their hyperdynamic circulation
advanced cirrhosis. Cystatin C concentrations are and decreased systemic vascular resistance and ascites is
Epidemiology, pathophysiology, and HRS management 23

Stage 1 AKI Stage 2 or 3 AKI

Close monitoring
Withdrawal of diuretics
Remove or minimise risk factors (remove and volume expansion
NSAIDs, minimise nephrotoxic drugs, with albumin*
reduce or withdraw diuretics, treatment of
infection if present)
Consider volume expansion with albumin*
response

Resolution Stable Progression


Yes No

Close follow up
Meet criteria for HRS

Further treatment of
AKI decided on a case- Yes No
by-case basis.

Vasoconstrictors Specific
(terlipressin or treatment
noradrenaline in for other
USA) and AKI
albumin** phenotypes

Figure 2. Management of different stages of AKI in cirrhosis. NSAIDs, nonsteroidal anti-inflammatory drugs. *Dose of albumin = 1 g/kg/d for
2 days. **Albumin should be continued at a dose of 20 to 40 g/d.

challenging.10 Unfortunately, monitoring central venous HRS AKI73 to counteract splanchnic arterial vasodila-
pressure has a limited role because it correlates poorly tion.74 The goal of using albumin is to combat the hemo-
with the intravascular response to fluid challenges. In dynamic dysfunction by antagonizing the decreased
addition, the presence of ascites leads to increased cen- effective circulating volume and increasing the mean
tral venous pressure without a correspondingly high ven- arterial pressure.
tricular preload.70 The most commonly used vasoconstrictors are norepi-
A meta-analysis of 38 reports comparing the use of nephrine, vasopressin analogues (terlipressin), somato-
hydroxyethyl starch, crystalloids, albumin, and gelatin in statin analogues (octreotide), and midodrine.75 The
intensive care patients with AKI showed that volume original studies were performed with vasopressin, but
resuscitation with albumin in comparison with crystal- because of ischemic side effects, other analogues were
loids does not reduce mortality, whereas hydroxyethyl developed. Terlipressin is the most extensively studied
starch was associated with a significantly increased risk vasopressor in this group,76 and now is considered as the
of AKI and mortality.71 Although another study showed first line of treatment of choice in treating HRS AKI
no significant difference in 90-day mortality after resus- patients in Europe. This is because it has a greater affin-
citation with 6% hydroxyethyl starch or saline, patients ity for vasopressin 1 receptors in the splanchnic bed
resuscitated with 6% hydroxyethyl starch were more compared with vasopressin 2 receptors in the kidneys,
likely to require renal replacement therapy, and, as such, thus predominantly exerting its vasoconstrictor effects
clinical guidelines recommend avoidance of synthetic on the splanchnic viscera without causing equal renal
starches in patients with cirrhosis.72 vasoconstriction.76 Even so, terlipressin is a powerful
vasoconstrictor and can cause ischemia, so should not be
used in patients with symptomatic ischemic heart dis-
Vasoconstrictors and Albumin ease, peripheral vascular disease, or recent stroke.
Clinical guidelines recommend using vasoconstrictors in Although vasopressin is available in the United States,
combination with albumin as the first-line treatment for terlipressin, which was developed more than 20 years
24 A.A. Amin et al.

ago to have greater vasopressin 1 specificity,77 is not cur- with antibiotic prophylaxis in advanced cirrhosis with
rently approved by the Food and Drug Administration low ascitic fluid protein, has been reported to prevent
for use in the United States or Canada because some HRS-AKI.88,89 Albumin not only has a role in maintain-
recent large randomized controlled trials failed to show a ing plasma oncotic pressure, and carrying trace elements
clear benefit for terlipressin in treating HRS type 1.78 and hormones, but also plays a major role in terms of
Numerous studies and meta-analyses have been con- detoxification. Fluid replacement with albumin has theo-
ducted to investigate the use of different vasopressors retical advantages over crystalloids in terms of its anti-
and albumin in managing HRS-AKI (Table 3).79,80 A inflammatory and antioxidant properties.88
network meta-analysis including 16 randomized con-
trolled clinical trials of patients with HRS reported that
Ascitic drainage
the combinations of terlipressin and albumin, and nor-
adrenaline and albumin, were more effective than albu- AKI resulting from abdominal compartment syndrome is
min therapy alone to achieve a complete reversal of well recognized when intra-abdominal pressure typically
HRS, as defined by a reduction in serum creatinine con- exceeds 18 mm Hg,89 although AKI can develop with
centration to less than 1.5 mg/d, whereas the combina- lower pressures.90 In patients with cirrhosis, paracentesis
tions of octreotide and albumin, or midodrine and is recommended for symptomatic relief.69 However,
albumin, failed to show any beneficial effects compared simple drainage of ascites can result in hypotension and
with albumin alone.81 However, there were no significant ischemic kidney injury, and, as such, replacement with
differences between these different regimens in terms of albumin is recommended for drainage of 5 L or more.
HRS AKI recurrence, other adverse events, or mortality, Uncontrolled studies have reported an improvement in
in keeping with previous meta-analyses.81 renal function in patients with HRS AKI after paracente-
Comparator studies have suggested that norepineph- sis for increased intra-abdominal pressure, with a reduc-
rine is an attractive alternative to terlipressin in the treat- tion in intrarenal pressure with improved diastolic
ment of HRS AKI, and may be associated with fewer perfusion, shown with color Doppler ultrasound.91 How-
adverse events,2,82,83 whereas other studies showed that ever, simply removing ascitic fluid, but maintaining
administering terlipressin by a continuous infusion intra-abdominal pressure, did not improve renal func-
instead of four intermittent boluses is better toler- tion,92 and, as such, most clinicians wait until ascites are
ated.76,84 A recent prospective study explored the safety tense or symptomatic before attempting drainage.93,94
and efficacy of a continuous terlipressin infusion at Liver transplantation (LT) is the best and most definitive
2 mg/d versus bolus dosing starting at 0.5 mg, adminis- treatment option for HRS AKI by curing end-stage liver
tered four times a day in patients with HRS AKI, and disease and subsequently reversing HRS AKI.95 The
reported significantly fewer adverse events in the contin- overall 1- and 5-year survival rates after LT in the setting
uous infusion group in comparison with intermittent of pretransplantation AKI have been reported as 77%
bolus administration.85 In preliminary studies, terlipres- and 69%, respectively.96 Pretransplantation renal func-
sin has been given to outpatients as a continuous infusion tion has been shown to be an important predictor of renal
at 2 mg/d and has been reported to be safe, well toler- dysfunction after LT, with a shorter duration of HRS
ated, and an effective option for the treatment of HRS AKI (<4 wk), with better outcomes and greater renal
AKI as a bridge to transplant.86,87 recovery.97 However, if the AKI episode persisted for
There are data supporting the efficacy of human albu- longer than 6 weeks, then patients were less likely to
min solution in the treatment of HRS AKI.88 The optimal recover renal function, and as such should be considered
dose of albumin used for HRS AKI treatment remains to for combined liver-kidney transplantation.44
be established, and the doses administered vary between A recent retrospective observational study from
reports. However, a recent meta-analysis including 19 Japan reported that preoperative renal dysfunction
clinical studies showed that the most important factor in with a cut-off glomerular filtration rate of less than
predicting a successful response to albumin therapy 40 mL/min/1.73 m2 at admission was an independent
appears to be the cumulative dose.35 This meta-analysis risk factor for 1-year survival for living-related trans-
observed a dose-response relationship between the plant recipients, and the postoperative 1-, 3-, and
amount of infused albumin and survival, with signifi- 5-year survival rate was significantly lower in patients
cantly improved survival with increasing 100-gram with preoperative HRS AKI than in patients without
increments, and expected 30-day survival rates among HRS.98 Although LT is the ideal option, it is not
patients receiving cumulative albumin doses of 200, 400, available for all patients in all countries.
and 600 g were 43.2%, 51.4%, and 59.0%, respectively,
independent of treatment duration, vasoconstrictor type,
or mean arterial pressure.35 Renal Replacement Therapy
Moreover, volume expansion using albumin for The effect of renal replacement therapy (RRT) remains
patients with SBP after large-volume paracentesis, and controversial in the management of HRS AKI, with
Epidemiology, pathophysiology, and HRS management
Table 3. Characteristics of Randomized Controlled Trials of Albumin and Vasoconstrictors for Treatment of HRS
Study Intervention Group Control Group Concomitant Therapy End Points Outcomes

Terlipressin versus placebo or control studies


Solanki et al,108 2003 Terlipressin 1 mg every 12 h for Placebo for 15 days; Albumin 20 g/d intrave- 15-day mortality, HRS Terlipressin group showed significant
15 days; n = 12 n = 12 nously; dopamine ≤4 mg/ reversal; and incidence of improvement in urine output, creati-
min for the first 24-48 h; side effects nine clearance, serum creatinine, and
fresh-frozen plasma until overall survival, and all survivors had
central venous pressure reversal of HRS
reaches the normal range
Neri et al,109 2008 Terlipressin 1 mg every 8 h for Albumin alone for Albumin 1 g/kg on the first 1- and 3-month mortality; Patients from terlipressin group showed
5 days followed by 0.5 mg every 15 days, n = 26 day followed by 20-40 g/d HRS reversal; and inci- a significant improvement in renal
8 h for 14 days; n = 26 dence of side effects function and probability of survival
Sanyal et al,110 2008 Terlipressin 1 mg/6 h up to Placebo for 14 days; Albumin 100 g on day 1 2- and 6-month mortality; Terlipressin was superior to placebo for
2 mg/6 h £ 14 days; n = 56 n = 56 followed by 25 g/d HRS reversal; HRS HRS reversal (34% versus 13%)
recurrence; and inci-
dence of side effects
Martin-Llahi et al,111 2008 Terlipressin 1 mg every 4 h up to Albumin alone for Albumin 1 g/kg on the first 3-month mortality; HRS Terlipressin was superior to placebo for
2 mg every 4 h for 15 days; 15 days; n = 18 day followed by 40 g/d reversal; HRS recur- HRS reversal (43.5% versus 8.7%)
n = 17 albumin rence; and incidence of
side effects
Zafar et al,112 2012 Terlipressin 1 mg every 4 h for Albumin alone for 7-10 Albumin 1 g/kg followed by 3-month mortality and HRS Terlipressin was superior to placebo for
7-10 days; n = 25 days; n = 25 20-40 g/d reversal HRS reversal (40% versus 8%)
13
Boyer et al, 2016 Terlipressin 1 mg every 6 h up to Placebo for 14 days; Albumin 20-40 g/d 3-month mortality; HRS HRS reversal was achieved in 23.7% in
2 mg every 6 h for 14 days; n = 99 reversal; HRS recur- terlipressin group versus 15.2% in
n = 97 rence; and incidence of placebo group
side effects
Noradrenaline versus terlipressin studies
Alessandria et al,113 2007 Noradrenaline 0.1 mg/kg/min up to Terlipressin 1 mg every Albumin 40-80 g/d 1-, 3-, and 6-month mortal- Reversal of HRS was observed in 7 of
0.7 mg/kg/min until HRS reversal 4 h up to 2 mg every 4 h ity; HRS reversal; HRS the 10 patients (70%) treated with
or for a maximum of 14 days; for 28 days until HRS recurrence; and inci- noradrenalin and in 10 of the 12
n=5 reversal or for a maxi- dence of side effects of patients (83%) treated with
mum of 14 days; n = 4 albumin terlipressin
Ghosh et al, 114 2013 Noradrenaline (dose and duration Terlipressin (dose and Albumin (dose not 3-month mortality; HRS Reversal of HRS was observed in 53%
not reported); n = 30 duration not reported); reported) reversal; and HRS treated with noradrenaline versus
n = 30 recurrence 57% treated with terlipressin
115
Sharma et al, 2008 Noradrenaline 0.5 mg/h up to Terlipressin 0.5 mg every Albumin 20-40 g/d 15-day mortality; HRS Each group achieved 50% HRS
3 mg/h for 15 days; n = 20 6 h up to 2 mg every 6 h reversal; and incidence of reversal
for 15 days; n = 20 side effects
116
Singh et al, 2012 Noradrenaline 0.5 mg/h up to Terlipressin 0.5 mg/6 h Albumin 20 g/d 1-month mortality; HRS HRS reversal could be achieved in
3 mg/h until HRS reversal or a up to 2 mg every 6 h reversal; HRS recur- 39.1% in terlipressin group versus
maximum of 14 days; n = 23 until HRS reversal or for rence; and incidence of 43.4% in patients in noradrenaline
a maximum of 14 days; side effects group
n = 23
Midodrine plus octreotide versus terlipressin studies
Cavallin et al,117 2015 Midodrine 7.5 mg/8 h up to Terlipressin 3 mg/24 h up Albumin 1 g/kg on first day 1- and 3-month mortality; Significantly higher rate of recovery of
12.5 mg/8 h orally plus octreo- to 12 mg/24 h until HRS followed by 20-40 g/d HRS reversal; incidence renal function in terlipressin group
tide 100 mg/8 h up to 200 mg/8 h reversal or a maximum of side effects (70.4% versus 28.6%)
subcutaneously until HRS rever- of 14 d (with albumin);
sal or a maximum of 14 days n = 27
(with albumin); n = 22
(continued on next page)

25
26 A.A. Amin et al.

similar short-term (30 d) and long-term (180 d) survival

treated with noradrenalin versus 75%


increased in both treatment groups
compared with non−RRT-treated patients, suggesting
Srivastava et al,118 2015 Dopamine 2 mg/kg/min plus furo- Terlipressin 0.5 mg every Albumin 20 g/d; cefotaxime 1-month mortality and inci- In HRS-I, significant improvement in
renal function at the end of 5 days

HRS reversal was observed in 73%

treated with midodrine-octreotide


that routine use of RRT may not be beneficial in HRS.99
The Acute Dialysis Quality Initiative group recom-
mended renal support for patients with HRS AKI only if
there was an acute potentially reversible event, or liver
transplantation planned, because of the lack of any evi-
dence for a major survival benefit for RRT in HRS AKI,
and that without an easily reversible precipitant or poten-
Outcomes

tial liver transplant, 3-month survival is marginal at


best.48,98
Transjugular intrahepatic portosystemic shunting
(TIPS) is also an option for treatment of HRS AKI, espe-
rence; and incidence of

cially in patients failing to respond to pharmacologic


3-month mortality; HRS
dence of side effects

reversal; HRS recur-

treatment, or with frequent relapses. TIPS increases the


effective renal blood flow by decreasing portal pressure
and redistributing regional vascular resistance.100 TIPS
side effects
End Points

has been shown to reduce the potent renal vasoconstric-


tor endothelin-1, and reduce intrarenal pressure and
increase diastolic blood flow.101 A meta-analysis
reported on the efficacy and safety of TIPS for the treat-
2 g intravenously every 8

ment of HRS AKI and showed that serum creatinine,


sodium, blood urea nitrogen, urinary sodium excretion,
Concomitant Therapy

and urine volume significantly improved after TIPS.


Midodrine orally 5 mg 3 times/d up Noradrenaline 0.1 mg/kg/ Albumin 20-60 g/d
h and lactulose

However, the higher incidence of hepatic encephalopa-


thy limits the standard use of TIPS as a routine therapeu-
tic option for HRS.102

Experimental Agents
min up to 0.7 mg/kg/min
until HRS reversal or a
maximum of 15 days;
6 h for 5 days; n = 20

There is a clinical need to develop agents to reverse


HRS AKI. Serelaxin (a recombinant human relaxin-2)
is a peptide with vasoprotective properties, has been
Control Group

studied in rat models of cirrhosis, and has shown an


albumin

increase in renal perfusion by reducing renal vascular


n=6

resistance.103 Studies to compare intravenous sere-


laxin infusion with terlipressin intravenous bolus in
every 8 h up to 200 mg every 8 h
semide 0.01 mg/kg/h for 5 days;

until HRS reversal or for a maxi-

cirrhotic patients with renal impairment have shown


to 15 mg 3 times/d plus octreo-

that serelaxin infusion increased the total renal arte-


tide subcutaneously 100 mg

rial blood flow by 65% from baseline, and was safe


Midodrine plus octreotide versus noradrenaline studies
Dopamine plus furosemide versus terlipressin studies

mum of 15 days; n = 9

and well tolerated, with no adverse effects related to


Abbreviations: HRS-1, hepato-renal syndrome 1.

systemic blood pressure or hepatic perfusion, whereas


Intervention Group

there was no significant change in renal arterial blood


flow with terlipressin.103
Nebivolol is a third-generation nonselective vasodi-
n = 20

lator b-blocker and has been studied in D-galactos-


amine−induced HRS in rats, where it was reported to
have anti-oxidant, anti-inflammatory, and anti-apopto-
tic properties, with renoprotective and hepatoprotec-
tive effects. This potentially makes nebivolol a
Table 3 (Continued)

promising drug with a possible effect in the preven-


2011−2012;

tion of HRS AKI or as an add-on medication in


Tavakkoli,119

patients with known HRS.104


Pentoxifylline has been advocated for the treatment of
2012
Study

alcoholic hepatitis, and potentially can reduce inflamma-


tion by decreasing proinflammatory cytokines, such as
Epidemiology, pathophysiology, and HRS management 27

tumor necrosis a. A preliminary, randomized, placebo- 11. Moreau R, Jalan R, Gines P, et al. Acute-on-chronic liver failure
controlled, clinical trial studied the safety of adding pen- is a distinct syndrome that develops in patients with acute
toxifylline to albumin with midodrine and octreotide in decompensation of cirrhosis. Gastroenterology. 2013;144:
1426-37. 1437.e1-9.
the treatment of HRS-1 and was shown to be safe, but 12. Jalan R, Yurdaydin C, Bajaj JS, et al. Toward an improved defi-
further large-scale prospective studies are needed to vali- nition of acute-on-chronic liver failure. Gastroenterology.
date the efficacy of pentoxifylline.105 2014;147:4-10.
Rifaximin has been shown to reduce the incidence of 13. Boyer TD, Sanyal AJ, Wong F, et al. Terlipressin plus albumin is
HRS AKI, and additional studies have investigated the more effective than albumin alone in improving renal function in
patients with cirrhosis and hepatorenal syndrome type 1.
effect of administration to patients with cirrhosis and Gastroenterology. 2016;150:1579-89. e2.
ascites, and these have shown that the overall blood urea 14. Wong F, Pappas SC, Boyer TD, et al. Terlipressin improves
nitrogen and serum creatinine concentrations were statis- renal function and reverses hepatorenal syndrome in patients
tically significantly lower in the rifaximin group com- with systemic inflammatory response syndrome. Clin Gastroen-
pared with the control group who received standard terol Hepatol. 2017;15:266-72. e1.
15. Piano S, Rosi S, Maresio G, et al. Evaluation of the Acute Kid-
medical care.106 Rifaximin also may decrease the plasma ney Injury Network criteria in hospitalized patients with cirrhosis
concentrations of interleukin 6, tumor necrosis factor-a, and ascites. J Hepatol. 2013;59:482-9.
and endotoxins, which play a crucial role in the develop- 16. Garcia-Tsao G, Parikh CR, Viola A. Acute kidney injury in cir-
ment of SBP and HRS AKI.107 rhosis. Hepatology. 2008;48:2064-77.
17. Fang J-T, Tsai M-H, Tian Y-C, et al. Outcome predictors and
new score of critically ill cirrhotic patients with acute renal fail-
CONCLUSIONS ure. Nephrol Dial Transplant. 2008;23:1961-9.
18. Hartleb M, Gutkowski K. Kidneys in chronic liver diseases.
In conclusion, the change in the classification of AKI and World J Gastroenterol. 2012;18:3035-49.
further advances in understanding the pathophysiological 19. Belcher JM, Garcia-Tsao G, Sanyal AJ, et al. Association of AKI
with mortality and complications in hospitalized patients with
basis of this syndrome is likely to lead to newer therapies
cirrhosis. Hepatology. 2013;57:753-62.
for this terrible complication of cirrhosis, which is asso- 20. Scott RA, Austin AS, Kolhe N V, McIntyre CW, Selby NM.
ciated with high mortality rates. Acute kidney injury is independently associated with death in
patients with cirrhosis. Frontline Gastroenterol. 2013;4:191-7.
21. Fede G, D’Amico G, Arvaniti V, et al. Renal failure and cirrho-
REFERENCES sis: a systematic review of mortality and prognosis. J Hepatol.
2012;56:810-8.
1. Angeli P, Gines P, Wong F, et al. Diagnosis and management of 22. D’Amico G, Garcia-Tsao G, Pagliaro L. Natural history and
acute kidney injury in patients with cirrhosis: revised consensus prognostic indicators of survival in cirrhosis: a systematic review
recommendations of the International Club of Ascites. J Hepatol. of 118 studies. J Hepatol. 2006;44:217-31.
2015;62:968-74. 23. Salerno F, Gerbes A, Gines P, Wong F, Arroyo V. Diagnosis,
2. Weil D, Levesque E, McPhail M, et al. Prognosis of cirrhotic prevention and treatment of hepatorenal syndrome in cirrhosis.
patients admitted to intensive care unit: a meta-analysis. Ann Gut. 2007;56:1310-8.
Intensive Care. 2017;7:33. 24. Francoz C, Glotz D, Moreau R, Durand F. The evaluation of
3. Gines P, Guevara M, Arroyo V, Rodes J. Hepatorenal syndrome. renal function and disease in patients with cirrhosis. J Hepatol.
Lancet. 2003;362:1819-27. 2010;52:605-13.
4. El-Naggar MM, Khalil el-SA, El-Daker MA, Salama MF. Bacte- 25. Mehta RL, Kellum JA, Shah S V, et al. Acute kidney injury net-
rial DNA and its consequences in patients with cirrhosis and cul- work: report of an initiative to improve outcomes in acute kidney
ture-negative, non-neutrocytic ascites. J Med Microbiol. injury. Crit Care. 2007;11:1-8.
2008;57:1533-8. 26. Tsien CD, Rabie R, Wong F. Acute kidney injury in decompen-
5. Sole C, Sola E, Morales-Ruiz M, et al. Characterization of sated cirrhosis. Gut. 2013;62:131-7.
inflammatory response in acute-on-chronic liver failure and rela- 27. Fagundes C, Barreto R, Guevara M, et al. A modified acute kid-
tionship with prognosis. Sci Rep. 2016;6:32341. ney injury classification for diagnosis and risk stratification of
6. Krag A, Bendtsen F, Henriksen JH, Møller S. Low cardiac impairment of kidney function in cirrhosis. J Hepatol.
output predicts development of hepatorenal syndrome and 2013;59:474-81.
survival in patients with cirrhosis and ascites. Gut. 28. Martin P-Y, Gines P, Schrier RW. Nitric oxide as a mediator of
2010;59:105-10. hemodynamic abnormalities and sodium and water retention in
7. Schneider AW, Kalk JF, Klein CP. Effect of losartan, an angio- cirrhosis. N Engl J Med. 1998;339:533-41.
tensin II receptor antagonist, on portal pressure in cirrhosis. 29. Gines P. Schrier R. , Gines P. Renal failure in cirrhosis. N Engl J
Hepatology. 1999;29:334-9. Med. 2009;361:1279-90.
8. Salerno F, Gerbes A, Gines P, Wong F, Arroyo V. Diagnosis, 30. Sola E, Gines P. Renal and circulatory dysfunction in cirrhosis:
prevention and treatment of hepatorenal syndrome in cirrhosis. current management and future perspectives. J Hepatol.
Postgrad Med J. 2007;84:662-70. 2010;53:1135-45.
9. Wong F, Nadim MK, Kellum JA, et al. Working Party proposal 31. Kastelan S, Ljubicic N, Kastelan Z, Ostojic R, Uravic M. The
for a revised classification system of renal dysfunction in patients role of duplex-doppler ultrasonography in the diagnosis of renal
with cirrhosis. Gut. 2011;60:702-9. dysfunction and hepatorenal syndrome in patients with liver cir-
10. Davenport A, Sheikh MF, Lamb E, Agarwal B, Jalan R. Acute rhosis. Hepatogastroenterology. 2004;51:1408-12.
kidney injury in acute-on-chronic liver failure: where does hepa- 32. Koppel MH, Coburn JW, Mims MM, Goldstein H, Boyle JD,
torenal syndrome fit? Kidney Int. 2017;92:1058-70. Rubini ME. Transplantation of cadaveric kidneys from patients
28 A.A. Amin et al.

with hepatorenal syndrome—evidence for the functional nature and improves survival in cirrhosis. Gastroenterology.
of renal failure in advanced liver disease. N Engl J Med. 2007;133:818-24.
1969;280:1367-71. 52. Dong T, Aronsohn A, Gautham Reddy K, Te HS. Rifaximin
33. Iwatsuki S, Popovtzer MM, Corman JL, et al. Recovery from decreases the incidence and severity of acute kidney injury and
“hepatorenal syndrome” after orthotopic liver transplantation. N hepatorenal syndrome in cirrhosis. Dig Dis Sci. 2016;61:3621-6.
Engl J Med. 1973;289:1155-9. 53. Piano S, Schmidt HH, Ariza X, et al. Association between grade
34. Kalambokis GN, Tsiakas I, Christaki M, et al. Systemic hemody- of acute on chronic liver failure and response to terlipressin and
namic response to terlipressin predicts development of hepatore- albumin in patients with hepatorenal syndrome. Clin Gastroen-
nal syndrome and survival in advanced cirrhosis. Eur J terol Hepatol. 2018. Epub ahead of print .
Gastroenterol Hepatol. 2018;30:659-67. 54. Van Slambrouck CM, Salem F, Meehan SM, Chang A. Bile cast
35. Salerno F, Navickis RJ, Wilkes MM. Albumin treatment regimen nephropathy is a common pathologic finding for kidney injury
for type 1 hepatorenal syndrome: a dose-response meta-analysis. associated with severe liver dysfunction. Kidney Int.
BMC Gastroenterol. 2015;15:167. 2013;84:192-7.
36. Fabrizi F, Dixit V, Martin P. Meta-analysis: terlipressin therapy 55. Krones E, Eller K, Pollheimer MJ, et al. Norursodeoxycholic
for the hepatorenal syndrome. Aliment Pharmacol Ther. acid ameliorates cholemic nephropathy in bile duct ligated mice.
2006;24:935-44. J Hepatol. 2017;67:110-9.
37. Serste T, Melot C, Francoz C, et al. Deleterious effects of beta- 56. Mehta G, Mookerjee RP, Sharma V, Jalan R. Systemic inflam-
blockers on survival in patients with cirrhosis and refractory mation is associated with increased intrahepatic resistance and
ascites. Hepatology. 2010;52:1017-22. mortality in alcohol-related acute-on-chronic liver failure. Liver
38. Mandorfer M, Bota S, Schwabl P, et al. Nonselective b blockers Int. 2015;35:724-34.
increase risk for hepatorenal syndrome and death in patients with 57. Davenport A, Agrawal B, Wright G, et al. Can non-invasive
cirrhosis and spontaneous bacterial peritonitis. Gastroenterology. measurements aid clinical assessment of volume in patients with
2014;146:1680-90. e1. cirrhosis? World J Hepatol. 2013;5:433-8.
39. Acevedo J, Fernandez J, Prado V, et al. Relative adrenal insuffi- 58. Ostermann M, Joannidis M. Acute kidney injury 2016: diagnosis
ciency in decompensated cirrhosis: relationship to short-term and diagnostic workup. Crit Care. 2016;20:299.
risk of severe sepsis, hepatorenal syndrome, and death. Hepatol- 59. Cobbaert CM, Baadenhuijsen H, Weykamp CW. Prime time for
ogy. 2013;58:1757-65. enzymatic creatinine methods in pediatrics. Clin Chem.
40. Theocharidou E, Krag A, Bendtsen F, Møller S, Burroughs AK. 2009;55:549-58.
Cardiac dysfunction in cirrhosis − does adrenal function play a 60. Carvounis CP, Nisar S, Guro-Razuman S. Significance of the
role? A hypothesis. Liver Int. 2012;32:1327-32. fractional excretion of urea in the differential diagnosis of acute
41. Weichselbaum L, Gustot T. The organs in acute-on-chronic liver renal failure. Kidney Int. 2002;62:2223-9.
failure. Semin Liver Dis. 2016;36:174-80. 61. Patidar KR, Kang L, Bajaj JS, Carl D, Sanyal AJ. Fractional
42. Thabut D, Massard J, Gangloff A, et al. Model for end-stage liver excretion of urea: a simple tool for the differential diagnosis of
disease score and systemic inflammatory response are major acute kidney injury in cirrhosis. Hepatology. 2018;68:224-33.
prognostic factors in patients with cirrhosis and acute functional 62. Fenton RA, Knepper MA. Urea and renal function in the 21st
renal failure. Hepatology. 2007;46:1872-82. century: insights from knockout mice. J Am Soc Nephrol.
43. Navasa M, Follo A, Filella X, et al. Tumor necrosis factor and 2007;18:679-88.
interleukin-6 in spontaneous bacterial peritonitis in cirrhosis: 63. Stevens LA, Coresh J, Greene T, Levey AS. Assessing kidney
relationship with the development of renal impairment and mor- function—measured and estimated glomerular filtration rate. N
tality. Hepatology. 1998;27:1227-32. Engl J Med. 2006;354:2473-83.
44. Nadim MK, Kellum JA, Davenport A, et al. Hepatorenal syn- 64. Seo YS, Park SY, Kim MY, et al. Serum cystatin C level: an
drome: the 8th International Consensus Conference of the Acute excellent predictor of mortality in patients with cirrhotic ascites.
Dialysis Quality Initiative (ADQI) Group. Crit Care. 2012;16: J Gastroenterol Hepatol. 2018;33:910-7.
R23. 65. Belcher JM, Sanyal AJ, Peixoto AJ, et al. Kidney biomarkers and
45. Rodrıguez E, Elia C, Sola E, et al. Terlipressin and albumin for differential diagnosis of patients with cirrhosis and acute kidney
type-1 hepatorenal syndrome associated with sepsis. J Hepatol. injury. Hepatology. 2014;60:622-32.
2014;60:955-61. 66. Watany MM, Hagag RY, Okda HI. Circulating miR-21, miR-210
46. Barreto R, Fagundes C, Guevara M, et al. Type-1 hepatorenal and miR-146a as potential biomarkers to differentiate acute tubu-
syndrome associated with infections in cirrhosis: natural history, lar necrosis from hepatorenal syndrome in patients with liver cir-
outcome of kidney function, and survival. Hepatology. rhosis: a pilot study. Clin Chem Lab Med. 2018;56:739-47.
2014;59:1505-13. 67. Angeli P, Gines P, Wong F, et al. Erratum: diagnosis and man-
47. Claria J, Stauber RE, Coenraad MJ, et al. Systemic inflammation agement of acute kidney injury in patients with cirrhosis: revised
in decompensated cirrhosis: characterization and role in acute- consensus recommendations of the International Club of Ascites.
on-chronic liver failure. Hepatology. 2016;64:1249-64. J Hepatol. 2015;62:968-74.
48. Trawale JM, Paradis V, Rautou PE, et al. The spectrum of renal 68. Kalafateli M, Wickham F, Burniston M, et al. Development and
lesions in patients with cirrhosis: a clinicopathological study. validation of a mathematical equation to estimate glomerular fil-
Liver Int. 2010;30:725-32. tration rate in cirrhosis: the royal free hospital cirrhosis glomeru-
49. Shah N, Dhar D, El Zahraa Mohammed F, et al. Prevention of lar filtration rate. Hepatology. 2017;65:582-91.
acute kidney injury in a rodent model of cirrhosis following 69. Piano S, Tonon M, Angeli P. Management of ascites and hepa-
selective gut decontamination is associated with reduced renal torenal syndrome. Hepatol Int. 2018;12 Suppl 1:122-34.
TLR4 expression. J Hepatol. 2012;56:1047-53. 70. Marik PE, Baram M, Vahid B. Does central venous pressure pre-
50. Jo SK, Cha DR, Cho WY, et al. Inflammatory cytokines and dict fluid responsiveness? A systematic review of the literature
lipopolysaccharide induce fas-mediated apoptosis in renal tubu- and the tale of seven mares. Chest. 2008;134:172-8.
lar cells. Nephron. 2002;91:406-15. 71. Zarychanski R, Abou-Setta AM, Turgeon AF, et al. Association
51. Fernandez J, Navasa M, Planas R, et al. Primary prophylaxis of of hydroxyethyl starch administration with mortality and acute
spontaneous bacterial peritonitis delays hepatorenal syndrome kidney injury in critically ill patients requiring volume
Epidemiology, pathophysiology, and HRS management 29

resuscitation: a systematic review and meta-analysis. JAMA. 90. Shear W, Rosner MH. Acute kidney dysfunction secondary to the
2013;309:678-88. abdominal compartment syndrome. J Nephrol. 2006;19:556-65.
72. Myburgh JA, Finfer S, Bellomo R, et al. Hydroxyethyl starch or 91. Dalfino L, Tullo L, Donadio I, et al. Intra-abdominal hyperten-
saline for fluid resuscitation in intensive care. N Engl J Med. sion and acute renal failure in critically ill patients. Intensive
2012;367:1901-11. Care Med. 2008;34:707-13.
73. Colle I, Laterre P-F. Hepatorenal syndrome: the clinical impact 92. Umgelter A, Reindl W, Franzen M, Lenhardt C, Huber W,
of vasoactive therapy. Expert Rev Gastroenterol Hepatol. Schmid RM. Renal resistive index and renal function before and
2018;12:173-88. after paracentesis in patients with hepatorenal syndrome and
74. Arroyo V, Garcia-Martinez R, Salvatella X. Human serum albu- tense ascites. Intensive Care Med. 2009;35:152-6.
min, systemic inflammation, and cirrhosis. J Hepatol. 93. Savino JA, Cerabona T, Agarwal N, et al. Manipulation of ascitic
2014;61:396-407. fluid pressure in cirrhotics to optimize hemodynamic and renal
75. Gines P, Angeli P, Lenz K, et al. EASL clinical practice guide- function. Ann Surg. 1988;208:504-11.
lines on the management of ascites, spontaneous bacterial perito- 94. Davenport A, Ahmad J, Al-Khafaji A, Kellum JA, Genyk YS,
nitis, and hepatorenal syndrome in cirrhosis. J Hepatol. Nadim MK. Medical management of hepatorenal syndrome.
2010;53:397-417. Nephrol Dial Transplant. 2012;27:34-41.
76. Gifford FJ, Morling JR, Fallowfield JA. Systematic review with 95. Boyer TD, Sanyal AJ, Garcia-Tsao G, et al. Impact of liver trans-
meta-analysis: vasoactive drugs for the treatment of hepatorenal plantation on the survival of patients treated for hepatorenal syn-
syndrome type 1. Aliment Pharmacol Ther. 2017;45:593-603. drome type 1. Liver Transpl. 2011;17:1328-32.
77. Bernadich C, Bandi JC, Melin P, Bosch J. Effects of F-180, a 96. Ruiz R, Barri YM, Jennings LW, et al. Hepatorenal syndrome: a
new selective vasoconstrictor peptide, compared with terlipres- proposal for kidney after liver transplantation (KALT). Liver
sin and vasopressin on systemic and splanchnic hemodynamics Transpl. 2007;13:838-43.
in a rat model of portal hypertension. Hepatology. 1998;27: 97. Wong F, Leung W, Al Beshir M, Marquez M, Renner EL. Out-
351-6. comes of patients with cirrhosis and hepatorenal syndrome type 1
78. Papaluca T, Gow P. Terlipressin: current and emerging indica- treated with liver transplantation. Liver Transpl. 2015;21:300-7.
tions in chronic liver disease. J Gastroenterol Hepatol. 98. Okamura Y, Hata K, Inamoto O, et al. Influence of hepatorenal
2018;33:591-8. syndrome on outcome of living donor liver transplantation: a sin-
79. Sanyal AJ, Boyer TD, Frederick RT, et al. Reversal of hepatore- gle-center experience in 357 patients. Hepatol Res. 2017;47:
nal syndrome type 1 with terlipressin plus albumin vs. placebo 425-34.
plus albumin in a pooled analysis of the OT-0401 and REVERSE 99. Zhang Z, Maddukuri G, Jaipaul N, Cai CX. Role of renal
randomised clinical studies. Aliment Pharmacol Ther. replacement therapy in patients with type 1 hepatorenal syn-
2017;45:1390-402. drome receiving combination treatment of vasoconstrictor plus
80. Facciorusso A, Chandar AK, Murad MH, et al. Comparative effi- albumin. J Crit Care. 2015;30:969-74.
cacy of pharmacological strategies for management of type 1 100. Rossle M. TIPS: 25 years later. J Hepatol. 2013;59:1081-93.
hepatorenal syndrome: a systematic review and network meta- 101. Umgelter A, Reindl W, Geisler F, Saugel B, Huber W, Berger H,
analysis. Lancet Gastroenterol Hepatol. 2017;2:94-102. et al. Effects of TIPS on global end-diastolic volume and cardiac
81. Sridharan K, Sivaramakrishnan G. Vasoactive agents for hepa- output and renal resistive index in ICU patients with advanced
torenal syndrome: a mixed treatment comparison network meta- alcoholic cirrhosis. Ann Hepatol. 2010;9:40-5.
analysis and trial sequential analysis of randomized clinical tri- 102. Song T, R€ ossle M, He F, Liu F, Guo X, Qi X. Transjugular intra-
als. J Gen Intern Med. 2018;33:97-102. hepatic portosystemic shunt for hepatorenal syndrome: a system-
82. Zheng J-N, Han Y-J, Zou T-T, et al. Comparative efficacy of atic review and meta-analysis. Dig Liver Dis. 2018;50:323-30.
vasoconstrictor therapies for type 1 hepatorenal syndrome: a net- 103. Snowdon VK, Lachlan NJ, Hoy AM, et al. Serelaxin as a poten-
work meta-analysis. Expert Rev Gastroenterol Hepatol. tial treatment for renal dysfunction in cirrhosis: preclinical eval-
2017;11:1009-18. uation and results of a randomized phase 2 trial. PLoS Med.
83. Nanda A, Reddy R, Safraz H, Salameh H, Singal AK. Pharmaco- 2017;14:e1002248.
logical therapies for hepatorenal syndrome. J Clin Gastroenterol. 104. Atwa A, Hegazy R, Mohsen R, Yassin N, Kenawy S. Protective
2018;52:360-7. effects of the third generation vasodilatory beta - blocker nebivo-
84. Fabrizi F, Aghemo A, Messa P. Hepatorenal syndrome and novel lol against D-galactosamine - induced hepatorenal syndrome in
advances in its management. Kidney Blood Press Res. rats. Open Access Maced J Med Sci. 2017;5:880-92.
2013;37:588-601. 105. Stine JG, Wang J, Cornella SL, et al. Treatment of type-1 hepa-
85. Cavallin M, Piano S, Romano A, et al. Terlipressin given by con- torenal syndrome with pentoxifylline: a randomized placebo
tinuous intravenous infusion versus intravenous boluses in the controlled clinical trial. Ann Hepatol. 2018;17:288-94.
treatment of hepatorenal syndrome: a randomized controlled 106. Ibrahim ES, Alsebaey A, Zaghla H, Moawad Abdelmageed S,
study. Hepatology. 2016;63:983-92. Gameel K, Abdelsameea E. Long-term rifaximin therapy as a
86. Robertson M, Majumdar A, Garrett K, Rumler G, Gow P, Testro primary prevention of hepatorenal syndrome. Eur J Gastroenterol
A. Continuous outpatient terlipressin infusion for hepatorenal Hepatol. 2017;29:1247-50.
syndrome as a bridge to successful liver transplantation. 107. Kang DJ, Kakiyama G, Betrapally NS, Herzog J, Nittono H,
Hepatology. 2014;60:2125-6. Hylemon PB, et al. Rifaximin exerts beneficial effects indepen-
87. Vasudevan A, Ardalan Z, Gow P, Angus P, Testro A. Efficacy of dent of its ability to alter microbiota composition. Clin Transl
outpatient continuous terlipressin infusions for hepatorenal syn- Gastroenterol. 2016;7:e187.
drome. Hepatology. 2016;64:316-8. 108. Solanki P, Chawla A, Garg R, Gupta R, Jain M, Sarin SK. Bene-
88. Valerio C, Theocharidou E, Davenport A, Agarwal B. Human ficial effects of terlipressin in hepatorenal syndrome: a prospec-
albumin solution for patients with cirrhosis and acute on chronic tive, randomized placebo-controlled clinical trial. J
liver failure: beyond simple volume expansion. World J Hepatol. Gastroenterol Hepatol. 2003;18:152-6.
2016;8:345-54. 109. Neri S, Pulvirenti D, Malaguarnera M, et al. Terlipressin and
89. Acevedo JG, Cramp ME. Hepatorenal syndrome: update on albumin in patients with cirrhosis and type I hepatorenal syn-
diagnosis and therapy. World J Hepatol. 2017;9:293-9. drome. Dig Dis Sci. 2008;53:830-5.
30 A.A. Amin et al.

110. Sanyal AJ, Boyer T, Garcia-Tsao G, et al. A randomized, prospec- in the treatment of type 1 hepatorenal syndrome and predictors
tive, double-blind, placebo-controlled trial of terlipressin for type of response. Am J Gastroenterol. 2008;103:1689-97.
1 hepatorenal syndrome. Gastroenterology. 2008;134:1360-8. 116. Singh V, Ghosh S, Singh B, et al. Noradrenaline vs. terlipressin
111. Martın-Llahı M, Pepin MN, Guevara M, et al. Terlipressin and in the treatment of hepatorenal syndrome: a randomized study. J
albumin vs albumin in patients with cirrhosis and hepatorenal syn- Hepatol. 2012;56:1293-8.
drome: a randomized study. Gastroenterology. 2008;134:1352-9. 117. Cavallin M, Kamath PS, Merli M, et al. Terlipressin plus albu-
112. Zafar S, Haque I, Tayyab G, Khan G, Chaudry N. Role of terli- min versus midodrine and octreotide plus albumin in the treat-
pressin and albumin combination versus albumin alone in hepa- ment of hepatorenal syndrome: a randomized trial. Hepatology.
torenal syndrome. Am J Gastroenterol. 2012;107:S175-6. 2015;62:567-74.
113. Alessandria C, Ottobrelli A, Debernardi-Venon W, et al. Nor- 118. Srivastava S, Shalimar, Vishnubhatla S, et al. Randomized con-
adrenalin vs terlipressin in patients with hepatorenal syndrome: a trolled trial comparing the efficacy of terlipressin and albumin
prospective, randomized, unblinded, pilot study. J Hepatol. with a combination of concurrent dopamine, furosemide, and
2007;47:499-505. albumin in hepatorenal syndrome. J Clin Exp Hepatol.
114. Ghosh S, Choudhary NS, Sharma AK, et al. Noradrenaline vs 2015;5:276-85.
terlipressin in the treatment of type 2 hepatorenal syndrome: a 119. Tavakkoli H, Yazdanpanah K, Mansourian M. Noradrenalin ver-
randomized pilot study. Liver Int. 2013;33:1187-93. sus the combination of midodrine and octreotide in patients with
115. Sharma P, Kumar A, Shrama BC, Sarin SK. An open label, pilot, hepatorenal syndrome: randomized clinical trial. Int J Prev Med.
randomized controlled trial of noradrenaline versus terlipressin 2012;3:764-9.

Вам также может понравиться