18
Ther Adv Gastroenterol 2015, Vol. 8(2) 83–100 DOI: 10.1177/ 1756283X14564673 © The Author(s), 2014. Reprints and permissions: http://www.sagepub.co.uk/ journalsPermissions.nav Therapeutic Advances in Gastroenterology http://tag.sagepub.com 83 Introduction Ascites is the most common complication of cir- rhosis with attendant portal hypertension [Moore and Aithal, 2006]. The production of ascites is related to inadequate renal sodium excretion, which generates a positive sodium balance. A typ- ical finding in patients with cirrhosis and portal decompensation is arterial splanchnic vasodila- tion, which results in increased mesenteric blood flow. Arterial splanchnic vasodilation causes a decrease in effective blood volume, activating baro and volume receptors, the sympathetic nerv- ous system, and the renin–angiotensin–aldoster- one system (RAAS) and triggering the nonosmotic release of vasopressin. Increased renal sympa- thetic nerve activity leads to increased tubular sodium retention and a positive sodium balance. Renal sympathetic nerve activity is further enhanced by activation of the hepatorenal reflex secondary to increased sinusoidal pressure and/or a decrease in sinusoidal blood flow [Ming et al. 2002]. Increased vasodilation in the splanchnic area is counteracted by an increase in total blood flow, maintaining normal blood pressure and an adequate blood flow to all organs. However, fur- ther splanchnic vasodilation increases the activa- tion of vasoconstrictor systems and leads to central underfilling, which cannot be sufficiently counteracted by increasing cardiac output. This causes decreased blood flow to the kidney and prerenal failure, termed hepatorenal syndrome (HRS) in these patients. Ascites Ascites is a major complication of cirrhosis of the liver, and is mainly due to portal hypertension. Within 10 years of the diagnosis of cirrhosis, over Treatment and management of ascites and hepatorenal syndrome: an update Kurt Lenz, Robert Buder, Lisbeth Kapun and Martin Voglmayr Abstract: Ascites and renal dysfunction are frequent complications experienced by patients with cirrhosis of the liver. Ascites is the pathologic accumulation of fluid in the peritoneal cavity, and is one of the cardinal signs of portal hypertension. The diagnostic evaluation of ascites involves assessment of its granulocyte count and protein concentration to exclude complications such as infection or malignoma and to allow risk stratification for the development of spontaneous peritonitis. Although sodium restriction and diuretics remain the cornerstone of the management of ascites, many patients require additional therapy when they become refractory to this treatment. In this situation, the treatment of choice is repeated large-volume paracentesis. Alteration in splanchnic hemodynamics is one of the most important changes underlying the development of ascites. Further splanchnic dilation leads to changes in systemic hemodynamics, activating vasopressor agents and leading to decreased renal perfusion. Small alterations in renal function influence the prognosis, which depends on the cause of renal failure. Prerenal failure is evident in about 70% of patients, whereas in about 30% of patients the cause is hepatorenal syndrome (HRS), which is associated with a worse prognosis. Therefore, effective therapy is of great clinical importance. Recent data indicate that use of the new definition of acute kidney injury facilitates the identification and treatment of patients with renal insufficiency more rapidly than use of the current criteria for HRS. In this review article, we evaluate approaches to the management of patients with ascites and HRS. Keywords: acute kidney injury, ascites, diuretic therapy, hepatorenal syndrome Correspondence to: Kurt Lenz, MD Department of Internal and Intensive Care Medicine, Konventhospital Barmherzige Brüder Linz, Seilerstätte 2, Linz, A-4020, Austria kurt.lenz@meduniwien. ac.at Robert Buder, MD Martin Voglmayr, MD Department of Internal and Intensive Care Medicine, Konventhospital Barmherzige Brüder Linz, Austria Lisbeth Kapun, MD HFR Meyriez-Murten, Murten, Switzerland 564673TAG 0 0 10.1177/1756283X14564673Therapeutic Advances in GastroenterologyK. Lenz et al. review-article 2014 Review

Treatment and Management of Ascites And

Embed Size (px)

DESCRIPTION

tratamiento sindrome hepatorrenal

Citation preview

Ther Adv Gastroenterol

2015, Vol. 8(2) 83 –100

DOI: 10.1177/ 1756283X14564673

© The Author(s), 2014. Reprints and permissions: http://www.sagepub.co.uk/ journalsPermissions.nav

Therapeutic Advances in Gastroenterology

http://tag.sagepub.com 83

IntroductionAscites is the most common complication of cir-rhosis with attendant portal hypertension [Moore and Aithal, 2006]. The production of ascites is related to inadequate renal sodium excretion, which generates a positive sodium balance. A typ-ical finding in patients with cirrhosis and portal decompensation is arterial splanchnic vasodila-tion, which results in increased mesenteric blood flow. Arterial splanchnic vasodilation causes a decrease in effective blood volume, activating baro and volume receptors, the sympathetic nerv-ous system, and the renin–angiotensin–aldoster-one system (RAAS) and triggering the nonosmotic release of vasopressin. Increased renal sympa-thetic nerve activity leads to increased tubular sodium retention and a positive sodium balance. Renal sympathetic nerve activity is further enhanced by activation of the hepatorenal reflex

secondary to increased sinusoidal pressure and/or a decrease in sinusoidal blood flow [Ming et al. 2002]. Increased vasodilation in the splanchnic area is counteracted by an increase in total blood flow, maintaining normal blood pressure and an adequate blood flow to all organs. However, fur-ther splanchnic vasodilation increases the activa-tion of vasoconstrictor systems and leads to central underfilling, which cannot be sufficiently counteracted by increasing cardiac output. This causes decreased blood flow to the kidney and prerenal failure, termed hepatorenal syndrome (HRS) in these patients.

AscitesAscites is a major complication of cirrhosis of the liver, and is mainly due to portal hypertension. Within 10 years of the diagnosis of cirrhosis, over

Treatment and management of ascites and hepatorenal syndrome: an updateKurt Lenz, Robert Buder, Lisbeth Kapun and Martin Voglmayr

Abstract: Ascites and renal dysfunction are frequent complications experienced by patients with cirrhosis of the liver. Ascites is the pathologic accumulation of fluid in the peritoneal cavity, and is one of the cardinal signs of portal hypertension. The diagnostic evaluation of ascites involves assessment of its granulocyte count and protein concentration to exclude complications such as infection or malignoma and to allow risk stratification for the development of spontaneous peritonitis. Although sodium restriction and diuretics remain the cornerstone of the management of ascites, many patients require additional therapy when they become refractory to this treatment. In this situation, the treatment of choice is repeated large-volume paracentesis. Alteration in splanchnic hemodynamics is one of the most important changes underlying the development of ascites. Further splanchnic dilation leads to changes in systemic hemodynamics, activating vasopressor agents and leading to decreased renal perfusion. Small alterations in renal function influence the prognosis, which depends on the cause of renal failure. Prerenal failure is evident in about 70% of patients, whereas in about 30% of patients the cause is hepatorenal syndrome (HRS), which is associated with a worse prognosis. Therefore, effective therapy is of great clinical importance. Recent data indicate that use of the new definition of acute kidney injury facilitates the identification and treatment of patients with renal insufficiency more rapidly than use of the current criteria for HRS. In this review article, we evaluate approaches to the management of patients with ascites and HRS.

Keywords: acute kidney injury, ascites, diuretic therapy, hepatorenal syndrome

Correspondence to: Kurt Lenz, MD Department of Internal and Intensive Care Medicine, Konventhospital Barmherzige Brüder Linz, Seilerstätte 2, Linz, A-4020, Austria [email protected]

Robert Buder, MD Martin Voglmayr, MD Department of Internal and Intensive Care Medicine, Konventhospital Barmherzige Brüder Linz, Austria

Lisbeth Kapun, MD HFR Meyriez-Murten, Murten, Switzerland

564673 TAG0010.1177/1756283X14564673Therapeutic Advances in GastroenterologyK. Lenz et al.review-article2014

Review

Therapeutic Advances in Gastroenterology 8(2)

84 http://tag.sagepub.com

50% of patients develop ascites [Becker, 2011]. The development of ascites is associated with a poor prognosis and a mortality rate of 20% per year [D’Amico et  al. 2006]. Therefore, patients with ascites should be considered for liver trans-plantation, preferably before the development of renal dysfunction.

Because 15% of patients with cirrhosis of the liver develop ascites of nonhepatic origin, the cause of new-onset ascites must be evaluated in all patients by abdominal paracentesis. The analysis of ascitic fluid should include assessment of neutrophil count and total protein concentration. Inoculation of ascites into blood culture bottles should be per-formed at the bedside if infection of the ascitic fluid is suspected. A diagnosis of spontaneous bacterial peritonitis (SBP) is made in the presence of an ele-vated absolute polymorphonuclear leukocyte count (i.e. >250 cells/mm3) in the ascitic fluid without evi-dence of a surgically treatable intra-abdominal source of infection [Becker, 2011]. Determination of the ascitic protein concentration is necessary to identify patients at increased risk of the develop-ment of SBP: a protein concentration of <1.5 g/dl is a risk factor for development of the condition [Moore and Aithal, 2006]. In patients in whom a cause of ascites other than cirrhosis of the liver is suspected, determination of the serum–ascites albu-min gradient (SAAG) is useful. The SAAG is ⩾1.1 g/dl in ascites due to portal hypertension, with an accuracy of 97% [Gines et al. 2010]. If ascites associated with malignancy is suspected, positive results of cytologic analyses of ascitic fluid can be obtained in patients with peritoneal carcinomatosis, but not in those with liver metastasis or hepatoma [Runyon et al. 1988]. Paracentesis is considered a safe procedure even in patients with an abnormal prothrombin time, with an overall complication rate not exceeding 1%. More serious complications, such as bowel perforation or bleeding into the abdominal cavity, occur in fewer than 1 in 1000 paracenteses. Data supporting cutoff values for coagulation parameters are unavailable; routine prophylactic use of fresh-frozen plasma or platelets before paracentesis is therefore not recommended, although if thrombocytopenia is severe (platelet count ⩽40,000/μl), most clinicians would adminis-ter pooled platelets to reduce the risk of bleeding [Gines et al. 2010, Moore and Aithal, 2006].

TreatmentDecompensated cirrhosis due to chronic viral or autoimmune hepatitis often shows marked

improvement in response to antiviral or immuno-suppressive treatments, respectively. In most patients with alcoholic liver disease, abstinence from alcohol results in improvements in liver function and ascites [Becker, 2011]. There are no defined criteria for when the treatment of ascites should be initiated. Patients with clinically unap-parent ascites usually do not require specific ther-apy, but it is recommended that patients with clinically evident and symptomatic ascites undergo treatment [Moore et al. 2003].

Patients with ascites have a positive sodium bal-ance, i.e. sodium excretion is low relative to sodium intake. Hence, the mainstay of therapy for ascites is sodium restriction and diuretic ther-apy (Table 1). Sodium intake should be restricted to 5–6 g/day (83–100 mmol/day of NaCl). More stringent restriction is not recommended, because it may worsen the malnutrition often present in patients with cirrhosis [Soulsby et al. 1997]. Given that the spontaneous resolution of ascites is obtained by reducing dietary sodium content in only 10–20% of patients, a negative sodium balance cannot be achieved without the use of diuretics in most patients with cirrhosis and ascites. Activation of the RAAS in patients with cirrhosis of the liver causes hyperaldosteron-ism and increased reabsorption of sodium throughout the distal tubule [Bernardi et  al. 1985]. Therefore, aldosterone antagonists, such as spironolactone, or its active metabolite, potas-sium canrenoate [Perez-Ayuso et al. 1982], rep-resent first-line diuretics in the treatment of ascites in patients with cirrhosis. The initial dose is 100–200 mg/dl. Monotherapy with a loop diu-retic such as furosemide is less effective than use of spironolactone, and is not recommended (86) Perez-Ayuso et al. 1982. Two different schedules are used in clinical practice. The first (‘sequential diuretic treatment’) consists of the administra-tion of increasing doses of spironolactone. If the response to 200 mg spironolactone is insufficient within the first 2 weeks, furosemide is added at an initial dose of 20–40 mg/day. If necessary, the dose of spironolactone can be increased stepwise to a maximum of 400 mg/day, and the dose of furosemide can be increased to a maximum of 160 mg/day. The second schedule (‘combined diuretic treatment’) involves the simultaneous administration of an aldosterone antagonist and a loop diuretic from the beginning of treatment, and the dose of both diuretics can be increased if no response is achieved. An established scheme for initial combined therapy is 100 mg

K Lenz, R Buder et al.

http://tag.sagepub.com 85

spironolactone and 40 mg furosemide per day, administered in the morning. If this dosage is insufficient, a stepwise increase, keeping the spironolactone:furosemide ratio constant (e.g. 200 mg spironolactone:80 mg furosemide), is possible. In a study by Angeli and colleagues [Angeli et al. 2010], adverse effects (38% versus 20%; p < 0.05), in particular hyperkalemia (18% versus 4%; p  <  0.05), were more common in patients who received sequential therapy, and the percentage of patients whose ascites resolved without changing the effective diuretic step was higher with combined treatment (56% versus 76%; p  <  0.05). The combination of sodium restriction, spironolactone, and furosemide is sufficient therapy for ascites in 90% of patients with cirrhosis of the liver [Becker, 2011].

To avoid hypovolemia, daily weight loss in patients with ascites should not exceed 1000 g in the pres-ence of peripheral edema or 500 g in the absence of peripheral edema [Pockros and Reynolds, 1986]. The diuretic effect is sufficient when only small amounts of ascitic fluid remain and periph-eral edema has disappeared completely. Complications of diuretic therapy include hepatic encephalopathy, renal failure, gynecomastia, elec-trolyte disturbances such as hyponatremia and hypokalemia or hyperkalemia, and muscle cramps [Santos et al. 2003]. To minimize these complica-tions, it is advisable to reduce the dosage of diu-retic drugs after the mobilization of ascites. Complications of diuretic therapy are most fre-quent in the first few weeks of therapy. Amiloride is an alternative to spironolactone in patients with painful gynecomastia. It is administered at a dose of 10–40 mg/day, but it is less effective than potas-sium canrenoate [Angeli et  al. 1994]. Notably, hyponatremia, which is caused by impaired free

water excretion, itself reduces the effect of loop diuretics.

The level of hyponatremia at which diuretic treatment should be stopped is contentious. It is generally agreed that diuretics should be paused when serum sodium is <120–125 mmol/l [Runyon, 2012]. Low serum sodium concentra-tion is an independent predictor of mortality in patients with cirrhosis. The prevalence of low serum sodium concentration, as defined by serum sodium concentrations of <135 mmol/l, <130 mmol/l, <125 mmol/l, and <120 mmol/l, was found to be 49.4%, 21.6%, 5.7%, and 1.2%, respectively, in a large multicenter study involv-ing 994 patients with cirrhosis [Angeli et  al. 2006]. Given that total body sodium is usually not decreased in patients with ascites and hyponatremia (dilution hyponatremia = hyperv-olemic hyponatremia), fluid restriction is rec-ommended. Albumin infusion can be considered a treatment for hyponatremia in cirrhosis. The available data are limited and derive from a small number of patients with a short follow-up, but they suggest a benefit of albumin adminis-tration that warrants further exploration in larger randomized trials. A randomized pilot study of 24 patients admitted with serum sodium levels of <130 mmol/l found that the administration of albumin significantly improved serum sodium levels compared with matched controls treated with fluid restriction, generat-ing a mean increase of 9 mmol/l. There was also a significant increase, compared with controls, in free water clearance and a decrease in serum vasopressin levels in patients treated with albu-min [Jalan et al. 2007]. This suggests that albu-min contributes to the mitigation of circulatory dysfunction and decreases the nonosmotic

Table 1. Treatment of ascites.

Clinical findings Treatment

Ascites clinically not detectable No treatmentClinically evident ascites Combined treatment with:

• Dietary salt restriction • Aldosterone antagonists (starting dose of

spironolactone 50–100 mg tapering to a maximum of 400 mg until ascites is resolved)

• Loop diuretics (starting dose of furosemide 40 mg tapering to a maximum of 160 mg until ascites is resolved or hyponatremia occurs)

Ascites completely mobilized by diuretic treatment Reduce the diuretic dose by half every week under weight control

Therapeutic Advances in Gastroenterology 8(2)

86 http://tag.sagepub.com

release of arginine vasopressin [Gianotti and Cardenas, 2014; Umgelter et  al. 2012]. Hypervolemic hyponatremia should be differen-tiated from hypovolemic hyponatremia, a less common condition in cirrhosis in which hypov-olemia results from marked and prolonged loss of sodium. In this condition, if the hyponatremia is symptomatic, slow correction is indicated. The desirable rate of sodium increase is consid-ered to be 6–8 mEq/l in 24 hours, 12–14 mEq/l in 48 hours, and 14–16 mEq/l in 72 hours. A more rapid correction of serum sodium is not recommended, because of the risk of severe complications, especially pontine myelinolysis [Tzamaloukas et al. 2013].

In recent years, new drugs known as vaptans, which antagonize the vasopressin V2 receptor [Decaux and Vassart, 2008], have been used to treat hyponatremia caused by cardiac failure, syndrome of inappropriate antidiuretic hor-mone secretion (SIADH), and cirrhosis. In ran-domized studies, administration of tolvaptan induced a marked increase in urine flow and serum sodium concentration in patients with mild hyponatremia. However, in those with marked hyponatremia and renal failure, there was only a trend toward improvement of serum sodium concentration, and the effect only occurred during the administration of tolvap-tan. One week after vaptan cessation, hypona-tremia recurred [Cardenas et  al. 2012]. Tolvaptan was associated with an as yet unex-plained higher incidence of gastrointestinal bleeding when compared with the administra-tion of a placebo. Therefore, the use of tolvaptan warrants further long-term studies to evaluate its safety and efficacy.

Therapy for refractory and recurrent ascitesRefractory ascites is defined as ascites that does not respond to sodium restriction and high-dose diuretic treatment (400 mg/day spironolactone and 160 mg/day furosemide) or that recurs rap-idly after therapeutic paracentesis [Gines et  al. 2010]. About 5–10% of patients with cirrhosis and ascites are considered to have refractory ascites (63) (Krag et al. 2007). The median sur-vival of patients with ascites refractory to medical treatment is approximately 6 months. Recurrent ascites is defined as frequent admission to the hospital (more than three admissions per year) because of the reaccumulation of ascites [Arroyo, 2013].

Large-volume paracentesis (LVP) is the first-line therapy in patients with refractory ascites. In patients with recurrent ascites, the addition of vasopressors (with or without albumin) or albu-min alone can be tried, before invasive procedures such as paracentesis are attempted (Table 2). In patients with cirrhosis and activation of the sym-pathetic nervous system, the addition of clonidine to diuretic therapy induced an earlier diuretic response associated with reduced diuretic require-ments, fewer complications, and a lower incidence of readmission related to tense ascites [Laenerts et al. 1997]. Several mechanisms may contribute to the increased mobilization of ascites associated with the use of clonidine. Clonidine induced a reduction in norepinephrine concentration asso-ciated with an elevated glomerular filtration rate in patients with refractory ascites [Laenerts et al. 2006]. By increasing glomerular filtration rate and decreasing proximal reabsorption of sodium, clonidine may increase the delivery of sodium to the distal nephron, where spironolactone, initially less effective, increases natriuresis by impairing the distal reabsorption of sodium. In addition, clonidine decreases the portosystemic gradient, especially in patients with alcoholic cirrhosis.

Oral administration of midodrine is associated with a significant improvement in systemic hemo-dynamics in nonazotemic patients with cirrhosis and ascites. As a result, renal perfusion and renal sodium excretion also improve in these patients [Angeli et al. 1998]. In a randomized controlled trial, midodrine and a combination of clonidine, midodrine, and standard diuretic treatment con-trolled ascites significantly better than standard diuretic treatment alone over a 1-month period [Singh et al. 2013]. In a randomized trial involv-ing patients with refractory or recurrent ascites, oral midodrine at a dose of 7.5 mg three times daily was shown to increase urine volume, urinary sodium excretion, mean arterial pressure, and survival. Therefore, midodrine can used in addi-tion to diuretics to increase blood pressure and convert refractory ascites to diuretic-sensitive ascites [Singh et al. 2012a].

In a study by Krag and colleagues [Krag et  al. 2007] of 15 patients with nonrefractory ascites and 8 with refractory ascites, 2 mg of terlipressin increased glomerular filtration rate, sodium clear-ance, lithium clearance, osmolal clearance, und urinary sodium excretion, and decreased norepi-nephrine and plasma renin activity, compared with a placebo infusion. All parameters remained

K Lenz, R Buder et al.

http://tag.sagepub.com 87

unchanged after administration of the placebo. In addition, 2 mg of terlipressin increased water excretion during a water-loading test in nona-zotemic patients with cirrhosis (Child–Pugh classes B and C) but without hyponatremia [Krag et al. 2007; Kalambokis et al. 2010].

Albumin has been found to be effective for proph-ylaxis against ascites in small studies. This effect could be based on the finding of the reduced function of endogenous albumin in patients with liver failure [Garcia-Martinez et al. 2013]. A ran-domized unblinded trial showed that the long-term administration of diuretics plus human albumin (25 g/week in the first year and 25 g every 2 weeks thereafter) improved survival and reduced the risk of recurrence of ascites com-pared with the administration of diuretics alone [Romanelli et  al. 2006]. However, the relatively small sample size precluded any firm conclusions. In a randomized double-blind placebo-controlled trial, Bari and colleagues [Bari et al. 2012] found that albumin was not inferior to the combination of octreotide and midodrine in preventing the recurrence of ascites after LVP. In addition, the outcome was worse in patients given octreotide and midodrine.

Possible treatment options for refractory ascites include LVP, transjugular intrahepatic portosys-temic shunt (TIPS), and liver transplantation (Table 3). The first-line treatment for patients

with refractory ascites is LVP, wherein a needle is inserted into the peritoneal cavity to remove the ascitic fluid [Gines et al. 1987]. This procedure is easy to perform and safe in most instances. However, complications are not completely unknown, and may present after a delay [Martinet et  al. 2000]. Hemorrhagic complications are infrequent, despite the presence of coagulopathy in many patients. There are no data on the pro-phylactic use of fresh-frozen plasma or pooled platelets, except in patients with a very low plate-let count (<40,000/μl). However, LVP should be avoided in the presence of clinically overt dissem-inated intravascular coagulation. LVP larger than 5 l should be performed with the administration of albumin to decrease the risk of postparacente-sis circulatory dysfunction [Gines et  al. 2010]. The dose of albumin most commonly used is 8 g/l of ascitic fluid removed. However, lower doses may be effective, which could reduce the cost of the procedure. In an unblinded study, Allesandria and colleagues [Allesandria et  al. 2011] found that half the dose of albumin (4 g/l of ascitic fluid removed) was as effective as the standard dose in preventing paracentesis-induced circulatory dys-function and renal failure. However, the most important factor in avoiding paracentesis-induced circulatory dysfunction is to discontinue β-blocker therapy [Serste et al. 2010].

In selected patients, a TIPS is an alternative to serial LVP [Gines et  al. 2002]. This procedure

Table 2. Treatment of recurrent and refractory ascites.

Diagnosis Treatment

Recurrent ascites: frequent admission due to ascites reaccumulation (more than three per year)

• Oral clonidine 0.075 mg once to twice daily in alcoholic cirrhosis with systolic blood pressure ⩾135 mm Hg ± 20 g albumin/week in addition to sodium restriction and diuretic treatment

• Oral midodrine 75 mg three times daily ± 20 g albumin/week in addition to sodium restriction and diuretic treatment

• Intravenous terlipressin 2 mg daily ± 20 g albumin /week in addition to sodium restriction and diuretic treatment

• Large-volume paracentesis in nonresponders to clonidine or midodrine

• TIPS in nonresponders to clonidine or midodrineTherapy refractory ascites: no response to dietary restriction and diuretic treatment

• Large-volume paracentesis together with the administration of 8 g albumin/l ascitic fluid removed

• TIPS in nonresponders to large-volume paracentesis

TIPS, transjugular intrahepatic portosystemic shunt.

Therapeutic Advances in Gastroenterology 8(2)

88 http://tag.sagepub.com

creates a shunt between the portal and suprahe-patic veins, thereby reducing portal hypertension and the rate of recurrence of ascites. Use of a TIPS is associated with improved systemic hemo-dynamics and renal function. Many patients with refractory ascites regain the ability to excrete uri-nary sodium and can be maintained free of ascites without sodium restriction or diuretics. In con-trolled trials comparing paracentesis combined with intravenous albumin versus a TIPS in patients with cirrhosis and ascites, the TIPS was clearly worse, in terms of improvement of ascites and survival, in patients with refractory ascites than recurrent ascites [Gines et al. 2002; Lebrec et al. 1996; Rössle et al. 2000; Sanyal et al. 2003]. The most frequent complication of TIPS use is encephalopathy [Sanyal et al. 1994]; the incidence of new or worsening encephalopathy following TIPS use was found to be 20–31% [Salerno et al. 2004; Somberg et al. 1995]. In addition, a TIPS cannot be applied in some patients with advanced liver disease (Child–Pugh class C), congestive heart failure, and/or severe pulmonary hyperten-sion owing to an increased risk of mortality, or in patients with portal thrombosis or inadequate anatomy of the portal and hepatic veins. Given that TIPS use is not associated with an improve-ment in survival, total paracentesis with intrave-nous albumin for volume replacement is considered the treatment of choice for refractory ascites in cirrhosis.

During the past 50 years, several procedures aimed at improving the treatment of refractory ascites have been developed [Arroyo, 2013]. Peritoneovenous shunting (PVS) involves a multi-perforated intraperitoneal tube connected to a subcutaneously implanted unidirectional pump that permits the passage of fluid out of, but not into, the abdominal cavity, and a second subcuta-neous tube that reaches the superior vena cava through the internal jugular vein [LeVeen et  al. 1974]. When ascites is significant, it circulates

through the prosthesis in response to a pressure gradient (positive in the peritoneum, negative in the intrathoracic circulation). However, the flow decreases or even stops if central venous pressure increases, thus preventing fluid overload. Many studies have demonstrated that PVS is associated with major improvements in circulatory and renal function and a better response to diuretics in patients with refractory ascites [Gines et al. 1991]. However, these beneficial effects did not result in a significant improvement in the long-term man-agement of patients or in a reduction in the total time spent in hospital during follow up, because of a very high rate of shunt obstruction requiring reoperation. Moreover, these complications were not prevented by a redesign of the valve (Denver shunt) or by insertion of a thromboresistant tita-nium tip in the vascular end of the prosthesis [Gines et al. 1995]. Recently, an automated sys-tem was developed to remove ascites from the peritoneal cavity into the urinary bladder, from which it can be eliminated by normal urination [Bellot et  al. 2013]. The pump system removed 90% of the ascitic fluid and significantly reduced the median number of LVP sessions per month in 40 patients with recurrent ascites. Larger studies are needed to confirm the safety and efficacy of this procedure.

Hepatorenal syndromeRenal failure is a serious complication in patients with advanced cirrhosis. It occurs in about 20% of patients hospitalized with decompensated cir-rhosis [Garcia-Tsao et  al. 2008; Moore, 2013]. Renal dysfunction is diagnosed based on levels of creatinine and on creatinine-based equations. However, a recent study [Francoz et  al. 2010] showed that creatinine-based formulae overesti-mate the true glomerular filtration rate, especially in patients with cirrhosis aged under 50 years and in those with ascites. In about 70% of patients, renal failure is caused by prerenal failure due to

Table 3. Diagnostic criteria of HRS [Salerno et al. 2007].

Cirrhosis with ascites Serum creatinine > 1.5 mg/dl No improvement of serum creatinine (decrease to a level ⩽ 1.5 mg/dl) after at least 2 days of diuretic

withdrawal and volume expansion with albumin. The recommended dose of albumin is 1 g/kg of body weight per day up to a maximum of 100 g/day

Absence of shock No current or recent treatment with nephrotoxic drugs Absence of parenchymal kidney disease as indicated by proteinuria > 500 mg/day, microhematuria

(> 50 red blood cells per high-power field), and/or abnormal renal ultrasonography

K Lenz, R Buder et al.

http://tag.sagepub.com 89

gastrointestinal hemorrhage, bacterial infection, or hypovolemia (overuse of diuretics), and in about 30%, it is caused by intrarenal causes, e.g. hepatitis B- or C-associated glomerulonephritis or toxins. In about 70% of patients with prerenal failure, renal function can be restored with fluid replacement, but the remaining 30% are unre-sponsive to volume expansion [Garcia-Tsao et al. 2008].

HRS is a fully reversible impairment of renal function in patients with severe hepatic failure unresponsive to volume expansion. The preva-lence of HRS in patients affected by cirrhosis and ascites was found to be 18% after 1 year, increas-ing to 39% at 5 years [Gines and Schrier, 2009]. Before HRS is diagnosed, primary causes of renal failure, such as hypovolemia, infection, nephro-toxins, and other renal diseases, must be excluded. Nevertheless, in many patients with HRS lacking proteinuria or hematuria, some histologic changes are evident in kidney biopsies [Trawalé et al. 2010]. In almost half of patients with HRS, one or more precipitating factors were identified, including bacterial infections (57%), gastrointes-tinal hemorrhage (36%), and therapeutic para-centesis (7%) [Angeli et  al. 2010]. Although a precipitating event is often identifiable, HRS can also develop in an apparently spontaneous way. Spontaneous HRS causes no morphologic changes in the kidney, although morphologic alterations in the course of oligoanuria may reveal a transition from intermediate to irreversible kid-ney damage with necrosis of the tubules. In patients with HRS scheduled for liver transplan-tation, it may be prudent to consider simultane-ous kidney transplantation [Restuccia et  al. 2004].

HRS is diagnosed by excluding other forms of renal failure. The diagnostic criteria initially pro-posed by the International Ascites Club in 1996 were re-examined in 2007 [Salerno et  al. 2007] (see Table 4). Two types of HRS were identified.

Type 1, a rapid deterioration of renal function defined as an increase in serum creatinine to a value twice the baseline and >2.5 mg/dl within 2 weeks.

Type 2, a slow and gradual deterioration of renal function with serum creatinine >1.5 mg/dl, often in parallel to the worsening of cirrhosis and portal hypertension, and lead-ing to refractory ascites.

PathophysiologyThe reduced glomerular filtration rate evident in patients with HRS is due to poor renal circulation on the one hand and a reduced filtration fraction on the other. Several factors are considered to induce the impairment of renal function (Table 5).

Vasodilation in the splanchnic regionThis is considered the most important trigger of renal hypoperfusion. This vasodilation is associ-ated with enhanced formation of vasodilators and vascular hyporesponsiveness to vasocon-strictors. To date, several mechanisms have been identified that may contribute to this enhanced vasodilation and impaired vasoconstriction. Portal hypertension increases the production and release of endogenous vasodilators, such as nitric oxide, carbon monoxide, and cannabi-noids, into the splanchnic arterial circulation, but other factors seem to be involved as well. Even in the dormant phase of alcoholic liver dis-ease, vasoactive substances are produced in large quantities in the liver when the production of cytokines is increased. In the presence of endotoxin stimulation, such activation is increased disproportionately. Even in patients with nonalcoholic liver disease, infiltration of endotoxins may play a role. Thus, patients with HRS prior to liver transplantation exhibited sig-nificantly higher concentrations of interleukins 2 and 6 and tumor necrosis factor than patients lacking HRS before liver transplantation [Burke

Table 4. Diagnostic criteria for kidney dysfunction in cirrhosis using the acute kidney injury definition [Wong et al. 2011].

Diagnosis Definition

Acute kidney injury A rise in serum creatinine ⩾50% from baseline, or a rise of >0.3 mg/dlChronic kidney disease Glomerular filtration rate (GFR) <60 ml/min for >3 months calculated using the MDRD-6 formulaAcute on chronic kidney disease

A rise in serum creatinine ⩾50% from baseline or a rise of serum creatinine >0.3 mg/dl in a patient with cirrhosis whose GFR is <60 ml/min for >3 months, calculated using the MDRD-6 formula

MDRD-6 formula: GFR = 170 × serum creatinine−0.999 × age−0.176 × 1.180 (if black) × 0.762 (if female) × serum urea nitrogen−0.170 × Albumin0.138.

Therapeutic Advances in Gastroenterology 8(2)

90 http://tag.sagepub.com

et al. 1993]. Furthermore, increased endothelin concentrations have been observed, and regula-tory disorders of the adenosine system have also been discussed [Gerbes et  al. 1998]. Translocation of bacteria and bacterial products from the gut is likely to be a main factor under-lying the development of a chronic inflamma-tory state [Israelsen et  al. 2014]. Another mechanism seems to be defective contractile signaling in smooth muscle cells in response to vasoconstrictor stimulation [Hennenberg et  al. 2008]. In addition to the liver itself, little is known about the factors responsible for these defects. The consequences are vasodilation, increased plasma volume, and increased blood flow in the splanchnic circulation, leading to a reduction in the effective arterial blood volume and a drop in arterial pressure. Vasodilation is associated with increased mesenteric blood flow, which may in fact be a useful mechanism to maintain liver function. In one study, liver func-tion in patients who underwent transplantation for liver failure was found to be dependent on the in-vitro liver circulation [Cardoso et  al. 1994].

Central underfillingIn the early stages of cirrhosis, increased cardiac output compensates for the reduction in systemic vascular resistance caused by vasodilation in the splanchnic area, and allows arterial pressure and effective blood volume to remain within normal limits. In the advanced stages of cirrhosis, splanch-nic vasodilation is markedly increased, and addi-tional increases in cardiac output can no longer

compensate, leading to central underfilling. Central underfilling activates baro and volume receptors, with consequent stimulation of the sympathetic nervous system and RAAS and the nonosmotic release of vasopressin. This results in renal hypoperfusion caused by an increase in renal vascular resistance [Lenz et al. 1991], reab-sorption of sodium in the tubules, and increased reabsorption of water in the distal collecting tubules.

Abolished autoregulation of kidney perfusionIncreased sympathetic nervous system activity abolishes autoregulation in the kidney [Esler and Kye, 1998]. Consequently, the renal circulation becomes pressure-dependent, even in the pres-ence of physiologic blood pressure [Stadlbauer et  al. 2008]. This is because the mean arterial blood pressure at which renin starts to be released is shifted to <110 mmHg [Kirchheim and Ehmke, 1994; Persson et al. 1990].

Hepatorenal reflexIn cirrhosis, sinusoidal pressure is elevated by increased resistance and enhanced mesenteric blood flow. Elevated pressure, secondary to swell-ing in the course of glutamine infusion via the superior mesenteric artery, led to a reduction of renal circulation and the glomerular filtration rate. This was due to direct hepatorenal interac-tion [Lang et  al. 1991]. Increased intrahepatic pressure leads to enhanced activity of sympathetic efferents from the liver that, through synapses in the lumbar spinal cord, cause enhanced renal

Table 5. Pathomechanism of hepatorenal syndrome.

Underlying causes Consequences

Splanchnic vasodilation Central underfillingCentral underfilling Activation of volume- and baroreceptorsActivated volume and baroreceptors Increased sympathetic nerve activity, increased

RAAS, increased nonosmotic vasopressin releaseIncreased sympathetic activity, increased RAAS, increased nonosmotic vasopressin release

Renal vasoconstrictionAbolished autoregulation of the kidney

Increased intrahepatic pressure and decreased sinusoidal blood flow

Activation of the hepatorenal reflex, causing an increase in renal sympathetic nerve activity and a decrease in renal blood flow

Cardiac insufficiency Compensatory increase in cardiac output to maintain renal perfusion is lowered

Increased synthesis of vasoactive mediators Intrarenal vasoconstriction

RAAS, renin–angiotensin–aldosterone system.

K Lenz, R Buder et al.

http://tag.sagepub.com 91

sympathetic activity (the hepatorenal reflex [Kostreva et al. 1980]). A decrease in sinusoidal blood flow also triggers the hepatorenal reflex [Ming et al. 2002]. An increase in renal sympa-thetic nerve activity leads to renal vasoconstric-tion and increased tubular sodium reabsorption. Denervation of the kidney eliminates this renal sympathetic activity, thereby reducing the impair-ment of renal function. This can also be achieved by hepatic vagal denervation [Levy and Wexler, 1987]. In a clinical study, Solis-Herruzo and col-leagues [Solis-Herruzo et al. 1987] were able to interrupt the hepatorenal reflex with lumbar sym-pathetic block in patients with portal hyperten-sion and thereby improve renal function.

Cardiac insufficiencyAn increase in cardiac output is important to compensate for the reduction in systemic vascular resistance caused by vasodilation in the splanch-nic area, and permits the arterial pressure and effective blood volume to remain within normal limits. There is evidence that cardiac output decreases as cirrhosis progresses. Clinical, but not overt, cardiac insufficiency, caused by underlying viral diseases, toxins such as alcohol, and/or by cirrhotic cardiomyopathy per se, seems to be one of the leading risk factors for the development of HRS. Ruiz-del-Arbrol and colleagues [Ruiz-del-Arbrol et  al. 2005] showed that cardiac insuffi-ciency can be detected at an early stage in patients developing HRS. In addition, the use of β-blockers in patients with refractory ascites has been shown to be deleterious [Serste et al. 2010]. These find-ings suggest a cardiorenal link in advanced cir-rhosis (Krag et al. 2012b).

Relative adrenal insufficiencyThis is a common subclinical condition in patients with cirrhosis, and is diagnosed as an inadequate production of cortisol in response to stimulation with synthetic adrenocorticotropic hormone. Adrenal insufficiency increases the risk of bacte-rial infection, septic shock, and HRS [Acevedo et al. 2013]. Cortisol plays an important role in hemodynamic homeostasis: the response of the heart and blood vessels to catecholamines and angiotensin-II is increased by cortisol. Low levels of cortisol lead to an inadequate increase in car-diac output and insufficient vasoconstriction in response to stress. Accordingly, adrenal insuffi-ciency may be involved in the development of car-diomyopathy and HRS [Krag et al. 2012].

Treatment of renal failureTreatment should begin as soon as renal failure is diagnosed. The criteria for diagnosis of HRS may lead to renal failure being detected too late [Nadim et al. 2012]. In addition, creatinine-based formulae may overestimate the true glomerular filtration rate, because of the low levels of muscle proteins in patients with cirrhosis and ascites [Francoz et al. 2010]. Cystatin C-based equations have proven more accurate in the evaluation of renal function and the staging of chronic renal insufficiency [De Souza et al. 2014]. Use of the new definition of acute kidney injury (AKI) devel-oped by the Acute Kidney Injury Network as adapted for cirrhotic patients [Wong et al. 2011] (Table 6) allows renal failure to be identified much earlier [Moore, 2013]. A prospective obser-vational cohort study of patients with cirrhosis and AKI showed that AKI is frequently progres-sive and associated with mortality [Belcher et al. 2013]. Therefore, the early identification and treatment of its cause are the cornerstones of therapy (Table 6). This includes withdrawal of β-blockers in patients with end-stage cirrhosis [Krag et  al. 2012], discontinuation of nonsteri-odal drugs and diuretics and, in patients with hypovolemia due to bleeding, prompt treatment with fluids and blood products combined with measures to stop the bleeding [Baradarian et al. 2004].

To treat hypovolemia, the recommended dose of albumin is 1 g/kg body weight in all patients after at least 2 days of diuretic withdrawal [Salerno et  al. 2007]. However, because the degree of hypovolemia varies between patients, a more pre-cise estimation of the volume required appears logical. Indeed, using invasive instruments, Umgelter and colleagues [Umgelter et al. 2012] demonstrated that much more fluid (up to 2 g albumin/kg body weight) may be necessary to normalize renal function. The main problem in clinical practice is that invasive hemodynamic monitoring can only be performed in intensive care units. Nevertheless, if there are clinical signs that 1 g albumin/kg is inadequate, more fluid can be infused under the careful observation of non-invasive hemodynamic parameters and lung function.

Antiviral therapy may be effective in some patients with renal failure due to hepatitis C-related glo-merulonephritis after consideration of the possible adverse effects of this therapy in patients with advanced cirrhosis and hepatitis B virus-associated

Therapeutic Advances in Gastroenterology 8(2)

92 http://tag.sagepub.com

kidney injury [Gane et al. 2014]. Bacterial infec-tions must be identified and treated early. In patients with SBP, the addition of albumin (1.5 g/kg body weight at the time of diagnosis, followed by 1 g/kg on day 3) to antibiotic treatment reduces the incidence of renal failure [Salerno et al. 2013; Sort et al. 1999].

Ascites is a risk factor for the development of con-trast-induced nephropathy. Although no reports show that cirrhosis without ascites is a risk factor, all patients with cirrhosis who undergo radiologic studies and require contrast medium should receive standard prophylactic saline hydration [Lodhia et al. 2009]. A recent study [Mahmoodi et al. 2014] involved patients with coronary heart disease and chronic renal failure with a serum creatinine level of up to 4 mg/dl who were under-going coronary intervention, use of a bicarbonate solution (prepared by adding 154 ml of 1000 mEq/l sodium bicarbonate to 846 ml of 5% dex-trose in water) conferred greater protection than hydration with normal saline against contrast-induced AKI. However, because metabolic alka-losis, mostly due to low albumin concentration, is a frequent finding in liver failure [Funk et  al. 2006], further studies are required in patients with cirrhosis to confirm these results.

In end-stage cirrhosis, β-blockers may reduce survival owing to their negative impact on the car-diac compensatory reserve. The inability to increase cardiac output may compromise organ perfusion in these patients [Krag et al. 2012]. The use of β-blockers is associated with increased mortality in patients with refractory ascites at high risk of paracentesis-induced circulatory dys-function [Serste et  al. 2010]. Although data on this subject are scarce, the use of β-blockers should be avoided in patients with both cirrhosis and renal failure.

Pentoxifylline has been shown to reduce the risk of renal failure in patients with alcoholic hepatitis

in the short term, but this effect is no longer evi-dent after 6 months [Mathurin et al. 2013]. Oral antibiotics are effective in reducing the incidence of HRS in patients with advanced cirrhosis and ascites. In a randomized placebo-controlled study, primary prophylaxis with norfloxacin reduced the incidence of SBP and delayed the development of HRS in patients with cirrhosis, low-protein ascitic fluid (<15 g/l), and advanced liver failure (Child–Pugh score >9) [Fernandez et al. 2007].

Knowledge of the pathophysiologic features of HRS has led to a variety of therapeutic approaches. It would be logical to assume that renal function is improved by substances that abolish renal vaso-constriction. However, experiments performed with prostaglandin to date have produced diver-gent results [Wong et  al. 1994]. Likewise, the administration of dopamine achieves improve-ments in cirrhotic patients with relatively good renal function, but not in patients with massively impaired renal function [Bennet et al. 1975; Peschl, 1987]. In patients with cirrhosis, renal failure, and tense ascites, increased abdominal pressure (IAP) may contribute to renal dysfunction. Reduction of IAP following paracentesis and albumin substitu-tion improves renal function, probably by improv-ing renal blood flow [Umgelter et al. 2009).

The current therapeutic approach to HRS includes the administration of vasoconstrictive agents combined with albumin (Table 7). The effect of vasopressors on renal function in this situation is due to an increase in renal perfusion, resulting from increased central blood volume and elevated blood pressure. Given that autoregu-lation is diminished, increased blood pressure is associated with increased renal perfusion. An inadequate blood-pressure response to vasopres-sor therapy is a typical finding in nonresponders [Nazar et al. 2011; Velez and Nietert, 2011].

The available vasoconstrictors comprise vasopres-sin analogues and alpha-adrenergic agents such as

Table 6. Prophylaxis against hepatorenal system.

• Prompt and adequate treatment of hypovolemic situations • Albumin substitution in spontaneous bacterial peritonitis (SBP) • Albumin substitution with large-volume paracentesis • Antibiotic prophylaxis in patients at increased risk of SBP • Withdrawal of β-blockers in patients with cirrhosis with recurrent and refractory ascites • Withdrawal of nonsteroidal drugs in portal decompensation • Nephroprophylaxis in patients with cirrhosis when radiologic studies using contrast medium are

performed

K Lenz, R Buder et al.

http://tag.sagepub.com 93

noradrenaline and midodrine. The first studies were performed with octapressin, a vasopressin derivative, more than 40 years ago [Kew et  al. 1972]. Over 30 years ago, ornipressin (8-ornithine-vasopressin: a vasopressin derivative that predomi-nantly stimulates the vasopressin 1 receptor) was found to improve central hypovolemia through splanchnic vasoconstriction, leading to decreased activation of vasopressor hormones (norepineph-rine and renin) and decreased renal vascular resist-ance, resulting in improved renal perfusion in patients with cirrhosis and HRS [Lenz et al. 1991]. Because ornipressin was removed from the market, subsequent studies were performed using terlipres-sin. Terlipressin is a triglycyl-lysine-vasopressin with a vasoconstrictive effect per se when given as a bolus and an additional long-acting vasoconstric-tive effect caused by lysine-vasopressin, which is produced by triglycyl-lysine-vasopressin degrada-tion [Ryckwaert et al. 2009]. Vasopressors such as terlipressin induce vasoconstriction in the splanch-nic vasculature by acting on vasopressin 1 recep-tors, resulting in redistribution of blood flow to the kidneys. Vasopressin analogues have been shown to decrease serum levels of vasoconstrictor hormones such as norepinephrine and plasma renin activity, leading to a decrease in renal vascular resistance [Lenz et al. 1991]. The vasopressin analogue terli-pressin has been shown not only to improve renal function by peripheral actions, but also to signifi-cantly influence cardiac filling volumes [Krag et al. 2010]. In experimental ovine endotoxemia, inter-mittent bolus injections of terlipressin were linked

to decreased heart rate and cardiac index and increased pulmonary vascular resistance. Continuous infusion of terlipressin reversed endo-toxin-induced systemic arterial hypotension and improved left-ventricular stroke-work index [Lange et al. 2007]. Therefore, continuous administration of terlipressin may be superior to bolus injection.

Norepinephrine has a positive inotropic effect, which may be important in overcoming the car-diac insufficiency induced by chronic liver disease. In patients with portal hypertension, terlipressin increased thoracic and liver blood volumes and altered portal pressure by causing vasodilation of intrahepatic vessels [Kiszja-Kanowith et al. 2004].

The effect of terlipressin can be significantly enhanced by the addition of high doses of human albumin [Ortega et al. 2002]. Although the favora-ble effect of albumin in this context is tradition-ally linked to its role in volume replacement, other mechanisms related to its nononcotic properties are likely to be involved. Albumin binds to and carries a variety of hydrophilic and hydrophobic molecules, including metals, fatty acids, metabo-lites, and drugs. Many potentially toxic ligands are neutralized and catabolized by binding to albumin. Furthermore, albumin is the major source of extracellular reduced sulfhydryl groups, potent scavengers of reactive oxygen species gen-erated by oxidative stress; thus, albumin is the main circulating antioxidant in the body. Recent studies have shown that both albumin

Table 7. Treatment of hepatorenal syndrome.

Drug Dose

Terlipressin • Bolus: 0.5–2.0 mg intravenously every 4–6 hours, with stepwise dose increments if there is no improvement of serum creatinine, to a maximum of 12 mg/day or the occurrence of complications, in combination with albumin.

• Continuous infusion: 4 mg/day with stepwise dose increments if there is no increase in mean arterial blood pressure >10 mmHg or improvement in serum creatinine level, up to a maximum of 12 mg/day or the occurrence of complications, in combination with albumin.

Noradrenaline • Continuous infusion with a starting dose of 0.5 mg/h, with stepwise increments if there is no increase in mean arterial blood pressure >10 mmHg or improvement of serum creatinine level, up to a maximum of 3 mg/h or the occurrence of complications, in combination with albumin.

Midodrine plus octreotide • Oral midodrine 7.5–12.5 mg three times daily to increase mean arterial blood pressure >10 mmHg.

• Octreotide 200 μg subcutaneously three times daily.

Albumin • 1 g albumin/kg body weight on the first day, followed by 200–400 g daily.

Therapeutic Advances in Gastroenterology 8(2)

94 http://tag.sagepub.com

concentration and function are reduced in patients with cirrhosis [Jalan and Bernardi, 2013], and that the degree of albumin dysfunction is related to the prognosis of liver disease [Jalan and Bernardi, 2013; Oettl et  al. 2013]. In addition, albumin has been shown to have a positive car-diac inotropic effect in cirrhotic rats [Bortoluzzi et  al. 2013]. Substitution of albumin therefore seems to accomplish more than plasma expansion.

Many studies have been performed with terlipres-sin. Meta-analyses have demonstrated that the combination of terlipressin and albumin improves renal function in patients with HRS type 1 [Nazar et al. 2011; Neri et al. 2008] and reduces mortal-ity when compared with albumin alone. The reduction in mortality was evident in patients with type I, but not type II, HRS [Gluud et  al. 2010]. Therefore, terlipressin plus albumin may facilitate survival to transplantation and a better outcome after transplantation, because normali-zation of renal function before transplantation is associated with increased survival after transplan-tation [Restuccia et al. 2004]. In these studies, a fixed dose of 1 g albumin/kg body weight on the first day, with a reduction to 20–40 g on subse-quent days, was used. Terlipressin was started on the first day at a dose of 0.5 mg every 4–6 hours, with an increase to a maximum of 12 mg per day until improvement of renal function occurred, defined as a serum creatinine concentration of <1.25 mg/dl. An improvement was seen within 14 days [Martin-Llahi et  al. 2008; Neri et  al. 2008; Sanyal et al. 2008]. Predictors of improved renal function in patients with HRS include early stage renal failure, increased blood pressure induced by the vasopressor, normal sodium con-centration, and Child–Pugh score <11 [Boyer et al. 2011; Nazar et al. 2011; Velez and Nietert, 2011; Wong et  al. 2004]. Given that larger amounts of albumin-containing fluids have been shown to restore kidney function more effectively [Umgelter et  al. 2012], goal-oriented volume therapy, using serial echocardiography to assess changes in intravascular volume by measuring inferior vena caval diameter, right-ventricular end-diastolic volume index, left-ventricular end-diastolic area index, and global end-diastolic vol-ume index, may be useful [Charron et al. 2006], although studies on cirrhotic patients are scarce.

Also controversial are the parameters for titrating the dose of terlipressin. Given that a close rela-tionship exists between hemodynamics and the

improvement of renal function [Nazar et al. 2011; Velez and Nietert, 2011], an increase in mean arterial pressure of ⩾10 mmHg, as used in stud-ies with norepinephrine, may help to achieve an earlier improvement in renal function [Maddukuri et  al. 2014]. Adverse effects of terlipressin (pre-dominately ischemia) leading to the discontinua-tion of treatment occur in about 12% of patients [Fagundes and Gines, 2012]. Therefore, patients with coronary heart disease, peripheral vascular disease, and/or cerebrovascular disease should not be treated with terlipressin. Some of these complications may be caused by the bolus injec-tion, because of the acute effect of the prodrug [Ryckwaert et  al. 2009]. A continuous infusion may overcome this problem [Angeli et al. 2009]. Studies on endotoxic shock in animal models, in which continuous infusion of terlipressin exerted a superior effect on cardiac contractility than bolus injection [Lange et  al. 2007], support the advantages of continuous infusion of terlipressin.

Alpha-adrenergic agonists, such as norepineph-rine [Sharma et  al. 2008; Singh et  al. 2012] or midodrine, in combination with octreotide and albumin [Angeli et al. 1999; Skagen et al. 2009; Wong et al. 2004], are a reasonable alternative to terlipressin because of their low cost and wide availability. However, data on their use are lim-ited. They are recommended in the guidelines of the American Association for the Study of Liver Diseases (AASLD) because terlipressin is not yet available in the United States [Runyon, 2012], but they are not recommended by the European Association for the Study of the Liver (EASL) [Gines et al. 2010]. If HRS reoccurs after the dis-continuation of treatment, patients should be retreated with the same drug combination [Gines et al. 2010].

TIPS use was reported to be effective for HRS Type 1 in an uncontrolled study of a small group of patients [Guevara et al. 1998]. Insertion of a TIPS further improved renal function and sodium excretion in patients with HRS type 1, who responded to a combination of midodrine, octre-otide, and albumin [Wong et  al. 2004]. More studies are necessary to evaluate TIPS use in patients with HRS type 1. TIPS use is not recom-mended in the guidelines of the EASL or AASLD.

Type 2 HRS is a type of renal dysfunction that arises from the deterioration of several organ sys-tems, and is primarily caused by liver failure and portal hypertension. It is part of a gradually

K Lenz, R Buder et al.

http://tag.sagepub.com 95

developing multiorgan dysfunction, with central hypovolemia and cardiac dysfunction as the main pathogenic triggers [Lenz, 2005]. There are very few data about the use of vasopressors in combi-nation with albumin in patients with HRS type 2. The clinical picture is dominated by therapy for refractory ascites and hyponatremia. Therefore, repeated paracentesis is the first-choice treat-ment; if renal function is deteriorating, the use of vasopressors should be considered [Fagundes and Gines, 2012]. Patients must be evaluated to determine whether they are candidates for liver transplantation.

Renal replacement therapy (RRT) has been used in patients with type 1 HRS waiting for liver transplantation. There are no data comparing extracorporeal therapy with vasopressors plus albumin in patients with HRS; nevertheless, RRT is not considered to be a first-line treatment, because it does not correct the underlying patho-physiologic changes. RRT should be reserved for patients planned for liver transplantation. The mortality of patients with HRS treated with hemodialysis or hemofiltration in the intensive care unit is reportedly high [Witzke et al. 2004]. In a small study [Mitzner et al. 2000], improve-ment of renal function followed use of an extra-corporeal liver assist system (MARS®). However, in the recently published RELIEF trial, MARS therapy did not demonstrate a positive effect on patients with acute or chronic liver failure [Banares et al. 2013]. EASL guidelines state that RRT may be useful in patients who do not respond to vasoconstrictor therapy [Gines et  al. 2010]. It remains unclear what modality of RRT should be the first-choice treatment for pharma-cologic nonresponders awaiting transplantation.

Conflict of interest statementThe authors declare that they do not have any-thing to disclose regarding conflicts of interest with respect to this manuscript.

FundingThis research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

ReferencesAcevedo, J., Fernandez, J., Prado, V., Silva, A., Castro, M., Pavesi, M. et al. (2013) Relative adrenal insufficiency in decompensated cirrhosis: relationship

to short-term risk of severe sepsis, hepatorenal syndrome, and death. Hepatology 58: 1757–1765.

Allesandria, C., Ella, C., Mezzabitta, L., Risso, A., Andrealli, A., Spandre, M., Morgando, A. et al. (2011) Prevention of paracentesis-induced circulatory dysfunction in cirrhosis: Standard vs half albumin doses. A prospective, randomized, unblinded study. Digest Liver Dis 43: 881–886.

Angeli, P., Dalla Pria, M., De Bei, E., Albino, G., Caregaro, L., Merkel, C. et al. (1994) Randomized clinical study of the efficacy of amiloride and potassium canrenoate in nonacotemic cirrhotic patients with ascites. Hepatology 19: 72–79.

Angeli, P., Fasolato, S., Cavalllin, M., Trotta, E., Maresio, G., Callegaro, A. et al. (2009) Terlipressin given as continuous intravenous versus terlipressin given as intravenous boluses in the treatment of type 1 hepatorenal syndrome (HRS) in patients with cirrhosis. J Hepatol 50(Suppl. 1): S73.

Angeli, P., Fasolato, S., Mazza, E., Okolicsanyi, L., Maresio, G., Velo, E. et al. (2010) Combined versus sequential diuretic treatment of ascites in non-azotaemic patients with cirrhosis: results of an open randomised clinical trial. Gut 59: 98–104.

Angeli, P. and Morando, F. (2010) Optimal management of hepatorenal syndrome in patients with cirrhosis. Hepatic Med Evid Res 2: 87–98.

Angeli, P., Volpin, R., Gerunda, G., Maresio, G., Callegano, A., Galieto, A. et al. (1999) Reversal of type 1 hepatorenal syndrome (HRS) with the combined administration of midodrine and octreotide. Hepatology 29: 1690–1697.

Angeli, P., Volpin, R., Piovan, D., Bortoluzzi, A., Craighero, R., Bottaro, S. et al. (1998) Acute effects of the oral administration of midodrine, an alpha-adrenergic agonist, on renal hemodynamics and renal function in cirrhotic patients with ascites. Hepatology 28: 937–943.

Angeli, P., Wong, F., Watson, H. and Gines, P. (2006) Hyponatremia in cirrhosis: Results of a patient population survey. Hepatology 44: 1535–1542.

Arroyo, V. (2013) A new method for therapeutic paracentesis: The automated low flow pump system. Comments in the context of the history of paracentesis. J Hepatol 58: 850–852.

Banares, R., Nevens, F., Larsen, F., Jalan, R., Albillos, A., Dollinger, M. et al. (2013) Extracorporeal albumin dialysis with the molecular adsorbend recirculating system in acute-on-chronic liver failure. The RELIEF trial. Hepatology 57: 1153–1162.

Baradarian, R., Ramdhaney, S., Chapalamadugu, R., Skoczylas, L., Wang, K., Rivilis, S. et al. (2004) Early intensive resuscitation of patients with upper gastrointestinal bleeding decreases mortality. Am J Gastroent 9: 619–622.

Therapeutic Advances in Gastroenterology 8(2)

96 http://tag.sagepub.com

Bari, K., Minano, C., Shea, M., Inayat, I., Hasehm, H., Gilles, H. et al. (2012) The combination of octreotide and midodrine is not superior to albumin in preventin recurrence of ascites after large-volume paracentesis. Clin Gastroenterol Hepatol 10: 1169–1175.

Becker, E. (2011) Diagnosis and therapy of ascites in liver cirrhosis. World J Gastroenterol 17: 1237–1248.

Belcher, J., Garcia-Tsao, G., Sanyal, A., Bhogal, H., Lim, J., Ansar, N. et al. (2013) Association of AKI with mortality and complications in hospitalized patients with cirrhosis. Hepatology 57: 753–762.

Bellot, P., Welker, M., Soriano, G., von Schaewen, M., Appenrodt, B., Wiest, R. et al. (2013) Automated low flow pump systems for the treatment of refractory ascites: A multicenter safety and efficacy study. J Hepatol 58: 922–927.

Bennet, W., Keeffe, E., Melnyk, C., Mahler, D., Rösch, D., Rösch, J. et al. (1975) Response to dopamine hydrochloride in the hepatorenal syndrome. Arch Intern Med 135: 964–970.

Bernardi, M., Serfadeo, D., Trevisani, F., Rusticali, A. and Gasbarrini, G. (1985) Importance of plasma aldosterone concentration on the natriuretic effect of spironolactone in patients with liver cirrhosis and ascites. Digestion 31: 189–193.

Bortoluzzi, A., Ceolotto, G., Gola, E., Sticca, A, Bova, S., Morando, F. et al. (2013) Positive cardiac inotropic effect of albumin infusion in rodents with cirrhosis and ascites: molecular mechanisms. Hepatology 57: 266–276.

Boyer, T., Sanyal, A., Garcia-Tsao, G., Blei, A., Carl, D., Bexon, A. et al. (2011) Predictors of response to terlipressin plus albumin in hepatorenal syndrome (HRS) type 1: Relationship of serum creatinine to hemodynamics. J Hepatol 55: 315–321.

Burke, G., Cirocco, R., Roth, D., Fernandez, J., Allouche, M., Mardkou, M. et al. (1993) Activated cytokine pattern in hepatorenal syndrome: Fall in levels after successful orthotopic liver transplantation. Transplant Proc 25: 1876–1877.

Cardenas, A., Gines, P., Marotta, P., Czerwiec, F, Oyuang, J., Guevara, M. et al. (2012) Tolvaptan an oral vasopressin antagonist, in the treatment of hyponatremia in cirrhosis. J Hepatol 56: 571–578.

Cardoso, J., Gautreau, C., Jeyaraj, P., Patrzalek, D., Cherruau, B., Vauvourdolle, M. et al. (1994) Augmentation of portal blood flow improves function of human cirrhotic liver. Hepatology 19: 375–380.

Charron, C., Caille, V., Jardin, F. and Vieillard-Baron, A. (2006) Echocardiographic measurement of fluid responsiveness. Curr Opin Crit Care 12: 249–254.

D’Amico, G., Garcia-Tsao, G. and Pagliaro, L. (2006) Natural history and prognostic indicators of survival in cirrhosis. J Hepatol 44: 217–231.

Decaux, G. and Vassart, G. (2008) Non-peptide arginine vasopressin antagonists: the vaptanes. Lancet 371: 1624–1632.

De Souza, V., Hadj-Aissa, A., Dolomanova, O., Rabilloud, M., Rognant, N., Lemoine, S. et al. (2014) Creatinine-versus cystatine C-based equations in assessing the renal function of candidates for liver transplantation with cirrhosis. Hepatology 59: 1522–1531.

Esler, M. and Kye, D. (1998) Increased sympathetic nervous system activity and its therapeutic reduction in arterial hypertension, portal hypertension and heart failure. J Auton Nerv Syst 72: 210–219.

Fagundes, C. and Gines, P. (2012) Hepatorenal syndrome: A severe, but treatable cause of kidney failure in cirrhosis. Am J Kidney Dis 59: 874–885.

Fernandez, J., Navasa, M., Planas, R., Montoliu, S., Monfort, D., Doriano, G. et al. (2007) Primary prophylaxis of spontaneous bacterial peritonitis delays hepatorenal syndrome and improves survival in cirrhosis. Gastroenterology 133: 818–827.

Francoz, C., Prie, D., AbdelRazek, W., Moreau, R., Mandot, A., Belghitti, J. et al. (2010) Inaccuracies of creatinine and creatinine based equations in candidates for liver transplantation with low creatinine: Impact on the model for endstage liver disease score. Liver Transplant 16: 1169–1177.

Funk, G., Doberer, D., Fuhrmann, V., Holzinger, U., Kitzberger, R., Kneidinger, N. et al. (2006) The acidifying effect of lactate is neutralized by the alkalinizing effect of hypoalbuminemia in non-paracetamol-induced acute liver failure. J Hepatol 45: 387–392.

Gane, E., Deray, G., Liaw, Y., Lim, S., Lai, C., Rasenack, J. et al. (2014) Telbivudine improves renal function in patients with chronic hepatitis B. Gastroenterology 146: 138–146.

Garcia-Martinez, R., Caracenni, P., Bernardi, M., Gines, P., Arroyo, V. and Jalan, R. (2013) Albumin: Pathophysiologic basis of its role in the treatment of cirrhosis and its complications. Hepatology 58: 1836–1846.

Garcia-Tsao, G., Chirag, R. and Antonella, V. (2008) Acute kidney injury in cirrhosis. Hepatology 48: 2064–2077.

Gerbes, A., Gülber, V. and Bilzer, M. (1998) Endothelin and other mediators in the pathophysiology of portal hypertension. Digestion 59(Suppl. 2): 8–10.

Gianotti, R. and Cardenas, A. (2014) Hyponatraemia and cirrhosis. Gastroenterol Rep 2: 21–26.

K Lenz, R Buder et al.

http://tag.sagepub.com 97

Gines, P., Angeli, P., Lenz, K., Moller, S., Moore, K. and Moreau, R. (2010) EASL clinical practice guidelines on the management of ascites, spontaneous bacterial peritonitis, and hepatorenal syndrome in cirrhosis. J Hepatol 53: 397–417.

Gines, P., Arroyo, V., Quintero, E., Planas, R., Bory, F., Cabrera, J. et al. (1987) Comparison of paracentesis and diuretics in the treatment of cirrhotics with tense ascites: results of a randomized study. Gastroenterology 93: 234–241.

Gines, P., Arroyo, V., Vargas, V., Planas, R., Casafont, F., Panés, J. et al. (1991) Paracentesis with intravenous infusion of albumin as compared with peritoneovenous shunting in cirrhosis with refractory ascites. N Engl J Med 325: 829–835.

Gines, A., Planas, R., Angeli, P., Guarner, C., Salerno, F., Gines, P. et al. (1995) Treatment of patients with cirrhosis and refractory ascites by LeVeen shunt with titanium tip. Comparison with therapeutic paracentesis. Hepatology 22: 124–131.

Gines, P. and Schrier, R. (2009) Renal failure in cirrhosis. N Engl J Med 361: 1279–1290.

Gines, P., Uriz, J., Calahorra, B., Garcia-Tsao, G., Kamath, P., Ruiz-del-Arbol, L. et al. (2002) Transjugular intrahepatic portosystemic shunting versus paracentesis plus albumin for refractory ascites in cirrhosis. Gastroenterology 123: 1839–1847.

Gluud, L., Christensen, K., Christensen, E. and Krag, A. (2010) Systemic review of randomized trials of vasoconstrictor drugs for hepatorenal syndrome. Hepatology 51: 576–584.

Guevara, M., Gines, P., Bandi, C., Gilabert, R., Sort, P., Jimenez, W. et al. (1998) Transjugular intrahepatic portosystemic shunt in hepatorenal syndrome: effects on renal function and vasoactive systems. Hepatology 28: 416–422.

Hennenberg, M., Trebicka, J., Sauerbruch, T. and Heller, J. (2008) Mechanisms of extrahepatic vasodilation in portal hypertension. Gut 57: 1300–1314.

Israelsen, M., Gluud, L. and Krag, A. (2014) Acute kidney injury and hepatorenal syndrome in cirrhosis. J Gastroenterol Hepatol. DOI: 10.1111/jgh.12709.

Jalan, R. and Bernardi, M. (2013) Effective albumin concentration and cirrhosis mortality from concept to reality J Hepatol 59: 918–920

Jalan, R., Mookerjee, R., Cheshire, L., William, R. and Davies, N. (2007) Albumin infusion for severe hyponatremia in patients with refractory ascites: a randomized clinical trial. J Hepatol 46(Suppl. 1): S95.

Jalan, R., Schnurr, K., Mookerjee, R., Sen, S., Cheshire, L., Hodges, S. et al. (2009) Alterations in the functional capacity of albumin in patients with

decompensated cirrhosis is associated with increased mortality. Hepatology 50: 555–564.

Kalambokis, G., Pappas, K., Baltayiannis, G., Katsanou, A. and Tsianos, E. (2010) Effects of terlipressin on water excretion after oral water load test in nonazotemic cirrhotic patients with ascites without hyponatremia. Scand J Gastroenterol 45: 1509–1515.

Kew, M., Varma, R., Sampson, D. and Sherlock, S. (1972) The effect of octapressin on renal and intrarenal blood flow in cirrhosis of the liver. Gut 33: 14–20.

Kirchheim, H. and Ehmke, H. (1994) Vasoactive hormones: modulators of renal function. Clin Invest 72: 685–687.

Kiszja-Kanowith, M., Henriksen, J., Hansen, E., Moller, S. and Bendtsen, F. (2004) Effect of terlipressin on blood volume distribution in patients with cirrhosis. Scand J Gastroenterol 39: 486–492.

Kostreva, D., Castaner, A. and Kampine, J. (1980) Reflex effects of hepatic baroreceptors on renal and cardiac sympathetic nerve activity. Am J Physiol 238: R390–R394.

Krag, A., Bendtsen, F., Burroughs, A. and Moller, S. (2012) The cardiorenal link in advanced cirrhosis. Med Hypoth 79: 53–55.

Krag, A., Bendtsen, F., Mortensen, C., Henriksen, J. and Moller, S. (2010) Effects of a single terlipressin administration on cardiac function and perfusion in cirrhosis. Eur J Gastroenterol Hepatol 22: 1085–1092.

Krag, A., Moller, S., Henriksen, J., Holstein-Rathlou, N., Larsen, F. and Bendtsen, F. (2007) Terlipressin improves renal function in patients with cirrhosis and ascites without hepatorenal syndrome. Hepatology 46: 1863–1871.

Krag, A., Wiest, R., Albillos, A. and Gluud, L. (2012) The window hypothesis: haemodynamic and nonhaemodynamic effects of ß-blockers improve survival of patients with cirrhosis during a window in the disease. Gut 61: 967–969.

Laenaerts, A., Codden, T., Meunier, J., Henry, J. and Ligny, G. (2006) Effects of clonidine on diuretic response in ascitic patients with cirrhosis and activation of sympathetic nervous system. Hepatology 44: 844–849.

Laenerts, A., Coddon, T., Van Cauter, J., Moukhaiber, H., Meunier, J. and Ligny, G. (1997) Treatment of refractory ascites with clonidine and spironolactone. Gastroenterol Clin Biol 21: 524–525.

Lang, F., Tschernko, E., Schulze, E., Öttl, I., Ritter, M, Völkl, H. et al. (1991) Hepatorenal reflex regulating kidney function. Hepatology 14: 590–594.

Therapeutic Advances in Gastroenterology 8(2)

98 http://tag.sagepub.com

Lange, M., Morelli, A., Ertmer, C., Koehler, G., Bröking, K., Hucklenbruch, C. et al. (2007) Continuous versus bolus infusion of terlipressin in ovine endotoxemia. Shock 28: 623–629.

Lebrec, D., Giuily, N., Hadengue, A., Vilgrain, V., Moreau, R., Poynard, T. et al. (1996) Transjugular intrahepatic portosystemic shunts: comparison with paracentesis in patients with cirrhosis and refractory ascites: a randomized trial. J Hepatol 25: 135–144.

Lenz, K. (2005) Hepatorenal syndrome – is it hypovolemia, a cardiac disease, or part of gradually developing multiorgan dysfunction. Hepatology 42: 263–265.

Lenz, K., Hörtnagl, H., Druml, W., Reither, H., Schmid, R., Schneeweiss, B. et al. (1991) Ornipressin in the treatment of functional renal failure in decompensates liver cirrhosis. Gastroenterology 101: 1060–1067.

LeVeen, H., Christoudias, G., Ip, M., Luft, R., Flak, G. and Grosberg, S. (1974) Peritoneo-venous shunting for ascites. Ann Surg 180: 580–591.

Levy, M and Wexler, M. (1987) Hepatic denervation alters first phase urinary sodium excretion in dogs with cirrhosis. Am J Physiol 253: F664–F671.

Lodhia, N., Kader, M., Mayes, T., Mantry, P. and Maliakkal, B. (2009) Risk of contrast induced nephropathy in hospitalized patients with cirrhosis. World J Gastroenterol 15: 1459–1464.

Maddukuri, G., Cai, C., Munigala, S., Mohammadi, F. and Zhang, Z. (2014) Targeting an early and substantial increase in mean arterial pressure is critical in the management of type 1 hepatorenal syndrome: a combined retrospective and pilot study. Dig Dis Sci 59: 471–481.

Mahmoodi, K., Sohrabi, B., Ilkhchooyi, F., Malaki, M., Khaniani, M. and Hemmati, M. (2014). The efficacy of hydration with normal saline versus hydration with sodium bicarbonate in the prevention of contrast-induced nephropathy. Heart Views 15: 33–36.

Martin-Llahi, M., Pepin, M., Guevara, M., Diaz, F., Torre, A., Monescillo, A. et al. (2008) Terlipressin and albumin versus albumin in patients with cirrhosis and hepatorenal syndrome: a randomized study. Gastroenterology 134: 1352–1359.

Martinet, O., Reis, E. and Mosimann, F. (2000) Delayed hemoperitoneum following large-volume paracentesis in a patient with cirrhosis and ascites. Dig Dis Sci 45: 357–358.

Mathurin, P., Louvert, A., Duhamei, A., Nahon, P., Carbonell, N. Buursier, J. et al. (2013) Prednisolone with vs without pentoxifylline and survival of patients with severe alcoholic hepatitis. A randomized trial. JAMA 310: 1033–1041.

McCormick, P., Mistry, P., Kaye, G., Burroughs, A. and McIntyre, N. (1990) Intravenous albumin infusion is an effective therapy for hyponatraemia in cirrhotic patients with ascites. Gut 31: 204–207.

Mehta, R., Kellum, J., Shah, S., Molitoris, B., Ronco, C., Warnock, D. et al. (2007) Acute kidney injury network: report of an initiative to improve outcomes in kidney injury. Crit Care 11: R31.

Ming, Z., Smyth, D. and Lautt, W. (2002) Decreases in portal flow trigger a hepatorenal reflex to inhibit renal sodium and water excretion in rats: role of adenosine. Hepatology 35: 167–175.

Mitzner, S., Stange, J., Klammt, S., Risler, T., Erley, C., Bader, B. et al. (2000) Improvement of hepatorenal syndrome with extracorporeal albumin dialysis MARS: Results of a prospective, randomized, controlled trial. Liver Transplant 6: 277–286.

Moore, K. (2013) Acute kidney injury in cirrhosis: A changing spectrum. Hepatology 57: 435–437.

Moore, K. and Aithal, G. (2006) Guidelines on the management of ascites in cirrhosis. Gut 55(Suppl. 6): vi1–12.

Moore, K., Wong, F., Gines, P., Bernardi, M., Ochs, A. and Salerno, F. (2003) The management of ascites in cirrhosis: report on the consensus conference of the International Ascites Club. Hepatology 38: 258–266.

Nadim, M., Kellum, J., Davenport, A., Wong, F., Davis, C., Pannu, N. et al. (2012) Hepatorenal syndrome: the 8th international conference of the acute dialysis initiative (ADQI) group. Crit Care 16: R23.

Nazar, A., Pereira, G., Guevara, M., Martin-Llahi, M., Pepin, M., Marinelli, M. et al. (2011) Predictors of response to terlipressin plus albumin in hepatorenal syndrome (HRS) type 1: relationship of serum creatinine to hemodynamics. J Hepatol 55: 315–321.

Neri, S., Pulvirent, D., Malaguarnera, M., Cosimo, B., Bertino, G., Ignaccolo, L. et al. (2008) Terlipressin and albumin in patients with cirrhosis and type I hepatorenal syndrome. Dig Dis Sci 53: 830–835.

Oettl, K., Birner-Gruenberger, R., Spindelmoeck, W., Stueger, H., Dorn, L., Stadlbauer, V. et al. (2013). Oxidative albumin damage in chronic liver failure. A relation to albumin binding capacity, liver dysfunction and survival. J Hepatol 59: 978–983.

Ortega, R., Gines, P., Uriz, J., Cardenas, A., Calahorra, B., Las Heras, D. et al. (2002) Terlipressin therapy with and without albumin for patients with hepatorenal syndrome: Results of a prospective, nonrandomized controlled study. Hepatology 36: 941–948.

Perez-Ayuso, R., Arroyo, V., Planas, R., Gaya, J., Bory, F., Rimola, A. et al. (1982) Randomized

K Lenz, R Buder et al.

http://tag.sagepub.com 99

comparative study of efficacy of furosemide versus spironolactone in nonacezotemic cirrhosis with ascites. Relationship between the diuretic response and the activity of the renin–aldosterone system. Gastroenterology 84: 961–968.

Persson, P., Ehmke, H., Nafz, B. and Kirchheim, R. (1990) Sympathetic modulation of renal autoregulation by carotid occlusion in conscious dog. Am J Physiol 258: F364–F370.

Peschl, L. (1987) Klinische und experimentell Untersuchungen über die Wirkung von Dopamin auf die Hämodynamik und Funktion von Niere und Leber. Wien Klin Wochenschr 86(Suppl.): 1–33.

Pockros, P. and Reynolds, T. (1986) Rapid diuresis in patients with ascites from chronic liver disease: the importance of peripheral edema. Gastroenterology 90: 1827–1833.

Restuccia, T., Ortega, R., Guevara, M., Gines, P., Alessandria, C., Ozdogan, O. et al. (2004) Effects of treatment of hepatorenal syndrome before transplantation on posttransplantation outcome. A case control study. J Hepatol 40: 140–146.

Ripoli, C., Groszmann, R., Garcia-Tsao, G., Grace, N., Burroughs, A., Planas, R. et al. (2007) Hepatic venous gradient predicts clinical decompensation in patients with compensated cirrhosis. Gastroenterology 133: 481–488.

Romanelli, R., Villa, G., Barletta, G., Vizzuti, F., Lanini, F., Arena, U. et al. (2006) Long term albumin infusion improves survival in patients with cirrhosis and ascites: an unblinded randomized trial. World J Gastroenterol 12: 1403–1407.

Rössle, M., Ochs, A., Gulberg, V., Siegerstetter, V., Holl, J., Deibert, P. et al. (2000) A comparison of paracentesis and transjugular intrahepatic portosystemic shunting in patients with ascites. N Engl J Med 342: 1701–1707.

Ruiz-del-Arbrol, L., Nonescillo, A., Arocena, C., Valer, P., Gines, P., Moreira, V. et al. (2005) Circulatory function and hepatorenal syndrome in cirrhosis. Hepatology 42: 439–447.

Runyon, B. (2012) Introduction to the revised American Association for the Study of Liver Diseases practice guideline management of adult patients with ascites due to cirrhosis 2012. Hepatology 57: 1651–1653.

Runyon, B., Hoefs, J. and Morgan, T. (1988) Ascitic fluid analysis in malignancy- related ascites. Hepatology 8: 1104–1109.

Ryckwaert, F., Visolvy, A., Fort, A., Murat, B., Richard, S. Guillon, G. et al. (2009) Terlipressin, a provasopressin drug exhibits direct vasoconstrictor properties: Consequences on heart perfusion and performance. Crit Care Med 37: 876–881.

Salerno, F., Gerbes, A., Gines, P., Wong, F. and Arroyo, V. (2007) Diagnosis, prevention and treatment of hepatorenal syndrome in cirrhosis. Gut 56: 1310–1318.

Salerno, F., Merli, M., Riggio, O., Cazzaniga, M., Valeriano, V., Pozzi, M. et al. (2004) Randomized controlled study of TIPS versus paracentesis plus albumin in cirrhosis with severe ascites. Hepatology 40: 629–635.

Salerno, F., Navickis, R. and Wilkes, M. (2013) Albumin infusion improves outcomes of patients with spontaneous bacterial peritonitis: A metanalysis of randomized trials. Clin Gastroenterol Hepatol 11: 123–130.

Santos, J., Planas, R., Pardo, A., Durandez, R., Cabre, E., Morllas, R. et al. (2003) Spironolactone alone or in combination with furosemide in the treatment of moderate ascites in nonazotemic cirrhosis. A randomized comparative study of efficacy and safety. J Hepatol 39: 187–192.

Sanyal, A., Boyer, T., Garcia-Tsao, G., Regenstein, F., Rossaro, L., Appenrodt, B. et al. (2008) A randomized prospective double-blind placebo-controlled trial of terlipressin for type 1 hepatorenal syndrome. Gastroenterology 134: 1360–1368.

Sanyal, A., Freedman, A., Shiffman, M., Purdum, P., Luketic, V. and Cheatham, A. (1994) Portosystemic encephalopathy after transjugular intrahepatic portosystemic shunt: results of a prospective controlled study. Hepatology 20: 46–55.

Sanyal, A., Genning, C., Reddy, K., Wong, F., Kowdley, K., Benner, K. et al. (2003) The North America study for the treatment of refractory ascites. Gastroenterology 124: 634–641.

Serste, T., Francoz, C., Durand, F., Rautou, P., Valla, D., Moreau, R. et al. (2010) Deleterious effects of betablockers on survival in patients with cirrhosis and refractory ascites. Hepatology 52: 1017–1022.

Sharma, P., Kumar, A., Shrama, B. and Sarin, S. (2008) An open label, pilot, randomized controlled trial of noradrenaline versus terlipressin in the treatment of type 1 hepatorenal syndrome and predictors of response. Am J Gastroenterol 103: 1689–1697.

Singh, V., Dhungana, S., Singh, B., Vijayverghia, R., Nain, C., Sharma, N. et al. (2012a) Midodrine in patients with cirrhosis and refractory ascites: a randomized pilot study. J Hepatol 56: 348–354.

Singh, V., Ghosh, S., Singh, B., Kumar, P., Sharma, N. and Bhalla, A. (2012b) Noradrenaline vs. terlipressin in the treatment of hepatorenal syndrome: a randomized study. J Hepatol 56: 1293–1298.

Singh, V., Singh, A., Singh, B., Vijayvergiya, R., Sharma, N., Ghai, A. et al. (2013) Midodrine and

Therapeutic Advances in Gastroenterology 8(2)

100 http://tag.sagepub.com

clonidine in patients with cirrhosis and refractory or recurrent ascites: a randomized pilot study. Am J Gastroenterol 108: 560–567.

Skagen, C., Einstein, M., Lucey, M. and Said, A. (2009) Combination treatment with octreotide, midodrine, and albumin improves survival in patients with type 1 and type 2 hepatorenal syndrome. J Clin Gastroenterol 43: 680–685.

Solanki, P., Chawla, A., Garg, R., Gupta, R., Jain, M. and Sarin, S. (2003) Beneficial effects of terlipressin in hepatorenal syndrome: a prospective, randomized placebo-controlled clinical trial. J Gastroenterol Hepatol 18: 152–156.

Solis-Herruzo, J., Duran, A., Favela, V., Castellano, G., Madrid, J., Munoz-Yaguem, M. et al. (1987) Effects of lumbar sympathetic block on kidney function in cirrhotic patients with hepatorenal syndrome. J Hepatol 5: 167–173.

Somberg, K., Riegler, J., LaBerge, J., Doherty-Simor, M., Bachetti, P., Roberts, J. et al. (1995) Hepatic encephalopathy after transjugular intrahepatic portosystemic shunts: incidence and risk factors. Am J Gastroenterol 90: 549–555.

Sort, P., Navas, M., Arroyo, V., Aldeguer, X., Planas, R., Ruiz-del-Arbol, L. et al. (1999) Effect of intravenous albumin on renal impairment and mortality in patients with cirrhosis and spontaneous bacterial peritonitis. N Engl J Med 341: 403–409.

Soulsby, C., Madden, A. and Morgan, M. (1997) The effect of dietary sodium restriction on energy and protein intake. Hepatology 26(Suppl. 382A): 1013.

Stadlbauer, V., Wright, G., Banaji, M., Mukhopadhya, A., Mookejee, R., Moore, K. et al. (2008) Relationship between activation of the sympathetic nervous system and renal blood flow autoregulation in cirrhosis. Gastroenterology 134: 111–119.

Trawalé, J., Paradis, V., Rautou, P., Francoz, C., Escolano, S., Sallée, M. et al. (2010) The

spectrum of renal lesions in patients with cirrhosis: a clinicopathological study. Liver Int 30: 725–732.

Tzamaloukas, A., Malhotra, D., Rosen, B., Raj, D., Murata, G. and Shapiro, J. (2013) Principles of management of severe hyponatremia. J Am Heart Assoc 2(4): e000240.

Umgelter, A., Reindl, W., Huber, W. and Schmid, R. (2009) Renal resistive index and renal function before and after paracentesis in patients with hepatorenal syndrome and tense ascites. Intensive Care Med 35: 152–156.

Umgelter, A., Wagner, K., Reindl, W., Luppa, P., Geisler, F., Huber, W. et al. (2012) Renal and circulatory effects of large volume plasma expansion in patients with hepatorenals syndrome type 1. Ann Hepatology 11: 232–239.

Velez, J. and Nietert, P. (2011) Therapeutic response to vasoconstrictors in hepatorenal syndrome parallels increase in mean arterial pressure: a pooled analysis of clinical trials. Am J Kidney Dis 58: 928–938.

Witzke, O., Baumann, M., Patschan, D., Patschan, S., Mitchell, A., Treichel, U. et al. (2004) Which patients benefit from hemodialysis therapy in hepatorenal syndrome? J Gastroenterol Hepatol 19: 1369–1373.

Wong, F., Massie, D., Hsu, P. and Dudley, F. (1994) Dose dependent effects of oral misoprostol on renal function in alcoholic cirrhosis. Gastroenterology 106: 658–663.

Wong, F., Nadim, M., Kellum, J., Salerno, F., Bellomo, R., Gerbes, A. et al. (2011). Working party proposal for a revised classification system of renal dysfunction in patients with cirrhosis. Gut 60: 702–709.

Wong, F., Pantea, L. and Sniderman, K. (2004) Midodrine, octreotide, albumin and TIPS selected patients with cirrhosis and type 1 hepatorenal syndrome. Hepatology 40: 55–64.

Visit SAGE journals online http://tag.sagepub.com

SAGE journals