Topic Highlight Open Access
Copyright ©2010 Baishideng. All rights reserved.
World J Gastrointest Pharmacol Ther. Apr 6, 2010; 1(2): 54-63
Published online Apr 6, 2010. doi: 10.4292/wjgpt.v1.i2.54
Hepatic encephalopathy therapy: An overview
Oliviero Riggio, Lorenzo Ridola, Chiara Pasquale, Department of Clinical Medicine, Centre for the Diagnosis and Treatment of Portal Hypertension, “Sapienza” University of Rome, Rome 00185, Italy
Author contributions: All authors critically reviewed the literature, drafted the article and gave approval of the final version to be published.
Correspondence to: Oliviero Riggio, Professor, Department of Clinical Medicine, Centre for the Diagnosis and Treatment of Portal Hypertension, “Sapienza” University of Rome, Rome 00185, Italy. oliviero.riggio@uniroma1.it
Telephone: +39-6-49972001 Fax: +39-6-4453319
Received: December 15, 2009
Revised: January 24, 2010
Accepted: January 31, 2010
Published online: April 6, 2010

Abstract

Type-C hepatic encephalopathy (HE) is a severe complication of cirrhosis, which seriously affects quality of life and is strongly related to patient survival. Treatment based on a classical pharmacological approach that is aimed at reducing the production of gut-derived toxins, such as ammonia, is still under debate. Currently, results obtained from clinical trials do not support any specific treatment for HE and our competence in testing old and new treatment modalities by randomized controlled trials with appropriate clinically relevant end-points urgently needs to be improved. On the other hand, patients who are at risk for HE are now identifiable, based on studies on the natural history of the disease. Today, very few studies that are specifically aimed at establishing whether HE may be prevented are available or in progress. Recent studies have looked at non absorbable disaccharides or antibiotics and other treatment modalities, such as the modulation of intestinal flora. In the treatment of severe stage HE, artificial liver supports have been tested with initial positive results but more studies are needed.

Key Words: Complications of cirrhosis, Porto-systemic shunts, Ammonia, Nitrogen metabolism, Transjugular intrahepatic portosystemic shunt



INTRODUCTION

Hepatic encephalopathy (HE) is a neurological syndrome that occurs as a consequence of severe liver damage and portal hypertension. HE is divided into three types: (1) encephalopathy associated with acute liver failure [type-A (= acute) HE], occurring in patients with fulminant hepatitis; (2) encephalopathy associated with portal-systemic bypass [type-B (= bypass) HE], observed in patients with portal-systemic bypass and no intrinsic hepato-cellular disease; and (3) [type-C (= cirrhosis) HE][1] most frequently observed in patients with cirrhosis and portal hypertension. This review is on the treatment of the latter type of HE only.

In cirrhotic patients, HE may be clinically overt or minimal. The term minimal HE (MHE) includes a number of cognitive deficits such as alterations of psychomotor speed and executive functions[2], detectable in patients with liver cirrhosis only by psychometric[3-5] or electrophysiological techniques[6,7]. Clinically, overt HE (OHE) may be further divided into episodic (developing over a short period of time and fluctuating in severity), or persistent (with continuous neurological symptoms negatively affecting the patient’s self-sufficiency). Both episodic and persistent HE may be induced by a precipitating event or may occur apparently spontaneously[1]. Some HE precipitating events are: constipation, hypo- or hyperkalemia, alkalosis, hyponatremia, dietary indiscretion, hypovolemia, gastrointestinal bleeding, dehydration, infections, surgery, renal failure, anaemia, diuretics and psychoactive medications. Defining type-C HE into minimal or overt, episodic or persistent and precipitated or spontaneously occurring is clinically relevant since the management of each category is very different. Moreover, in planning clinical trials on HE treatment, including patients that are homogeneous according to the above classifications is of crucial importance.

Type-C HE represents a major clinical problem. In cirrhotic patients who were followed from a time when the disease was compensated, HE represents, in fact, the second most frequent cause of decompensation after ascites and before variceal bleeding[8]. HE is particularly frequent in patients undergoing portal-systemic shunt[9-11], and is considered an important prognostic factor for survival[12-15]. Both overt and MHE have a detrimental effect on the overall quality of life[15], since even MHE impairs the execution of simple and complex tasks, such as driving[16-18]. For these reasons, research searching for a better treatment is ongoing.

At present, many aspects of HE remain a matter of debate and are seen as “beyond treatment”. The current recommendations are based on the generic hypothesis that the symptoms are caused by the loss of a “protective” mechanism exerted by the liver on brain functions. As an effect of liver failure and porto-systemic shunting, substances arising from the gut are able to reach the systemic circulation and the central nervous system, where they can exert a “toxic effect” on brain function. This concept goes back to the beginning of the last century and is supported by: (1) the high incidence of HE after surgical, radiological or spontaneous porto-systemic shunting[9,10,19-22]; (2) the improvement of HE after bowel cleansing by enema[23] or gut irrigation[24]; (3) the improvement of HE by decreasing the stent diameter of transjugular intrahepatic porto-systemic shunts (TIPS)[22,25], and consequently, increasing the amount of intestinal blood shunted into the systemic circulation; and (4) the improvement of HE by closing large spontaneous porto-systemic shunts[26]. Any attempt to clarify the nature of the substances involved in the pathogenesis of HE, as well as the exact mechanism affecting brain function, have been unsatisfactory until now. Ammonia is still incriminated more often, but several other compounds, such as mercaptans, short-chain fatty acids amines, γ-aminobutyric acid (GABA), endorphins, glutamate, endogenous benzodiazepine agonists, tryptophan and several of its metabolites have also been investigated.

However, the role of these factors is still a matter of debate. As far as the mechanism involved in the central nervous system is concerned, in the last few years, astrocyte swelling has been identified as an important process negatively influencing neuronal neurotransmission as well as the brain energy production rate. Moreover, astrocyte swelling may affect the function of important brain proteins by causing oxidative and nitrosative stress with evidence of protein tyrosine nitration and, more recently, ribonucleic acid (RNA) oxidation[27]. The astrocyte swelling hypothesis is able to explain one of the key features of HE, namely that the syndrome is precipitated by heterogeneous factors. Many of these factors have been shown to induce the swelling of astrocytes by different mechanisms[28]. The infection may, for example, induce astrocyte swelling by endotoxins and pro-inflammatory cytokines[29]. Moreover, it has been recently proposed that neutrophils, in addition to ammonia, may be involved in the pathogenesis of HE. Thus, neutrophils can be a target for future anti-inflammatory therapeutic strategies in addition to ammonia lowering therapies[30].

Uncertainties on the pathogenesis of HE limit the development of specific pharmacological therapies. Nevertheless, treatments such non-absorbable disaccharides or antibiotics, gut cleansing by enemas or nasogastric tubes, general measures such as nutritional supports, correction of the precipitating events and of electrolyte imbalance have been used empirically for a long time and are considered standard treatments[31] in patients with HE. Unfortunately, their efficacy cannot be considered as “evidence-based”. In fact, most studies were performed before the era of rigorous randomized controlled trials (RCTs) and well-designed therapeutic trials on HE are warranted. It can be difficult to objectively stage HE’s severity and to appropriately select clinically relevant end-points[32].

PREVENTION OF HE

A number of observational studies on the natural history of HE and on risk factors for its development are in the literature. These studies allow the identification of patients at risk for developing this complication of liver cirrhosis with sufficient confidence. Therefore, prevention of HE is possible. Preventive treatments are above all needed by patients at high risk of HE, such as those with more advanced liver disease (Child C or ascites)[33], those undergoing radiological[9,11,19-22] or surgical porto-systemic shunts, as well as those bearing large spontaneous shunts in whom episodes of HE are very frequent or persistent[21]. Other patients likely to be selected for preventive measures are those who have already had at least one episode of HE in the past[34] and those with MHE at their first observation[4,35,36]. These two conditions are in fact associated with bouts of HE at follow up.

Given the large prevalence of patients at risk of HE, the possible advantages of a life- long preventive treatment should be balanced against possible adverse events and costs and should be tested by appropriate RCTs. These studies should be specifically aimed at establishing whether HE can be prevented. Their design should be different from that used in HE treatment studies. In fact, the ideal preventive study should include patients without HE at entry, and the main end-point should be the occurrence of any overt episode of HE during follow-up. This end point is very objective and easily identifiable. Each study should include patients with a given risk factor for HE. For example, all patients who survived a first episode of HE or, as another example, all patients submitted to a transjugular intrahepatic portosystemic shunt (TIPS) The sample size for the study could be estimated from the incidence of HE in the population at risk (for example, from the incidence of HE occurring in patients submitted to a TIPS). The inclusion of a “no-treatment” or a “placebo” group is mandatory. Cost/benefit ratios, as well as data on tolerability and safety, should be considered. Survival should be a secondary end point of the study. Since a prophylactic treatment should be prolonged life-long, the ideal therapy should be extremely safe and well tolerated.

In clinical practice, measures to reduce the nitrogenous load from the gut, such as long-term administration of non-absorbable disaccharides, are commonly applied in patients with advanced cirrhosis[37]. However, their efficacy in preventing HE has been specifically tested only very recently; one RCT report is now available on patients submitted to a TIPS and another has been reported on patients who recovered from an episode of OHE (secondary prophylaxis) (Table 1).

Table 1 RCTs on the prevention of episodes of overt hepatic encephalopathy.
Ref.Type and NO. of patients includedAim of the studyTested treatment (s)Control treatmentResults
Riggio et al[38] 2005Patients submitted to TIPS (75)Prevention of post TIPS HELactitol (60 g/d) rifaximin (1200 mg/d)No treatment (25)No difference between treatment and control groups
Sharma et al[39] 2009Patients who recovered from HE (140)Prevention of recurrence of HE (secondary prophylaxis)Lactulose (30-60 mL in 2 or 3 divided doses)No treatment (70)Lactulose effective
Kanematsu et al[42] 1988Patients submitted to surgery (56)Prevention of HE precipitated by surgeryBCAA enriched solution, (29)Conventional AA solution (27)No difference between treatment and control groups
Rolachon et al[24] 1994Patients bleeding from varicesPrevention of HE precipitated by bleedingGut cleansing using mannitol by naso-gastric tubeNo treatmentGut cleansing effective
Bass et al[53] 2009History of HEPrevention of recurrence of HE (secondary prophylaxis)Rifaximin 550 mg twice daily for 6 moPlaceboRifaximin effective

In post-TIPS patients, a recent RCT demonstrated that there was no difference in the incidence of OHE during the first month after TIPS placement, regardless of whether the patients received a prophylaxis with lactitol, rifaximin or no treatment[38]. On the contrary, lactulose has been shown to be able to significantly prevent the occurrence of a second episode of HE in patients who recovered from the first episode[39]. In both studies, a control group receiving no treatment was included. Therefore, low-absorbable disaccharides (lactitol or lactulose) resulted in effective prevention of recurrence of HE after a first episode but not in the prevention of HE after a TIPS. These different results underline the need for including homogeneous patients with specific risk factors in studies aimed at HE prophylaxis. In fact, HE in the first month after TIPS placement may be particularly difficult to prevent. Further compromise of first-pass hepatic clearance of ammonia is to be expected after TIPS placement. Additionally, splanchnic blood flow increases when there is a major reduction of the porto-systemic pressure gradient. Thus, delivery of ammonia to the systemic circulation may increase. Another factor to consider is upregulation of intestinal glutaminase activity, which has been reported to increase after porto-systemic shunt procedures[40]. This enzyme is responsible for the large amount of ammonia generated by the small intestine. Accordingly, one might anticipate that in the immediate aftermath of a TIPS procedure, more "intense" HE therapy might be needed to prevent overt episodes of HE.

Another RCT aimed at studying the effect of a probiotic yoghurt on the psychometric performance of cirrhotic patients with MHE showed that the episodes of OHE at follow up were significantly lower that those observed in the no-treatment arm of the study[41].

Concerning the prevention of precipitant induced HE, a series of specific treatments may be adopted, such as blood aspiration by a nasogastric tube, gut cleansing (by means of gut lavage, oral laxatives or enemas), parenteral/enteral nutrition (in case of bleedings or infections that may lead to a negative energy balance), diet (e.g. no proteins, low proteins, vegetable proteins) and specific drugs [e.g. branched-chain amino acids (BCAAs), ornithine aspartate, non-absorbable disaccharides, antibiotics]. However, until now, RCTs specifically aimed at testing these prophylactic approaches are very few. The only examples are a RCT showing that gut cleansing by means of a solution of mannitol can reduce the incidence of post-hemorrhagic HE[24] and another study on post-operative parenteral nutrition that showed no differences in the rate of occurrence of HE between patients treated with BCAAs-enriched and conventional amino acid solutions[42] (Table 1).

TREATMENT OF HE
MHE

The diagnosis and treatment of MHE are still active matters of discussion[43]. Computerized tests have been recently proposed (and validated) in patients with MHE[44,45] and may represent an amelioration of our diagnostic capacity. Most studies available in the literature have shown that several pharmacological approaches seemed to ameliorate patient psychometric performances. However, MHE affects quality of life and is a risk factor for the development of episodes of OHE[4,35,36]. In the last few years, it has been repeatedly suggested that it may seriously impair the driving capacity of cirrhotic patients[16,18], which can be evaluated by driving simulators[17].

The recent demonstration that lactulose in patients with MHE is able to induce an amelioration in the quality of life[46] raised a new prospective in the therapeutic approach of this clinical condition (Table 2). In fact, the demonstration that a given treatment can ameliorate patient psychometric performances, although obtained in RCTs (Table 2), is meaningless because MHE is by definition a subclinical condition. Future studies on the treatment of MHE should be aimed a ameliorating the clinical consequences of this alteration. In addition to quality of life, possible end-points in these studies could be the prevention of overt episodes of HE, which are particularly frequent in patients with MHE, and the improvement of skills such as driving capacity. Nevertheless, the modification of psychometric tests should not be chosen as the main end-point of the study, but as criteria to include comparable patients. Since a treatment of MHE should be prolonged life-long, the ideal therapy should be extremely safe and well tolerated. The modulation of intestinal bacterial flora can be a valid therapeutic approach for MHE.

Table 2 RCTs on the treatment of MHE.
Ref.Type of studyType of HE/patients (n)Tested treatmentControl treatmentResults
Prasad et al[46] 2007RCTMHE (61)Lactulose (30-60 mL of lactulose in 2 or 3 divided doses)No treatmentLactulose improved the quality of life
Bajaj et al[41] 2008Prospective randomized trialMHE (25)Probiotic yoghurtNo treatmentProbiotic improved the psychometric performance
Liu et al[73] 2004Double blind, randomized studyMHE (55)Bioactive, fermentable fibers and lactic acid bacteria Bioactive, fermentable fibersPlaceboSynbiotic treatment improved the psychometric performance and the Child-Turcotte-Pugh class
Malaguarnera et al[74] 2009Randomized studyMHE (125)Bifidobacterium + fructo-oligosaccharidesLactuloseBoth treatments improve blood ammonia and psychometric performance
Precipitant-induced episodic HE

The cornerstones of treatment for precipitant-induced episodic HE are the identification and treatment of the precipitating events and the general support of patients. In fact, the prevention of falls or body injuries in disorientated patients, the care of bladder and bowel functions, the care of intravenous lines, the monitoring of fluid balance, the monitoring of blood glycaemia and electrolytes (such as of arterial blood gases), the correction of acid/base disorders, blood pressure monitoring and the avoidance of aspiration pneumonia are the strategies commonly applied to support patients with episodic HE. The nutritional status of the patient is also considered to be useful. An energy intake of 35/40 kcal/kg body weight per day and a protein intake of 1.2/1.5 g/kg body weight per day are recommended. Energy should be provided by glucose and fat in a ratio of 50%-65%/35%-50% of non-protein calories, according to the european society of parenteral and enteral nutrition guidelines for nutrition in liver disease[47].

Moreover, in patients with severe HE (grades III to IV), solutions with an increased content of BCAAs and a reduced amount of aromatic amino acids can ameliorate neurological symptoms by ensuring, at the same time, an adequate protein intake.

The action of a well-recognized precipitating factor acts as a trigger in precipitant-induced HE. Multiple precipitating events may coexist in the same patient. Although not specifically evaluated, the identification and correction of the precipitating event is considered the first-line effort in patients with this type of HE. If the symptoms do not ameliorate once the identified precipitating event is resolved, a well-known clinical rule is to search for a second complication, for example, a superimposed infection in a patient who has recently bled. Some strategies that are commonly applied to stop precipitating events are the following: (1) in patients with HE induced by gastrointestinal hemorrhage, stop the bleeding with vasoactive drugs, an endoscopic therapy or an angiographic shunt (TIPS), correct the anemia with a blood transfusion and use a nasogastric tube to facilitate upper gastrointestinal cleansing; (2) infections (e.g. pulmonary, of the urinary tract, spontaneous bacterial peritonitis) should be promptly treated with appropriate antibiotic therapies; (3) constipation should be resolved by cathartic and/or bowel enema, electrolyte abnormalities by discontinuing diuretics and correcting hypo- or hyperkalemia; (4) deterioration of renal function should be corrected, if possible, by stopping diuretics, treating dehydration and discontinuing nephrotoxic drugs; and (5) if HE is precipitated by the administration of exogenous sedatives, benzodiazepines should be discontinued and flumazenil, the competitive benzodiazepine antagonist that binds with the benzodiazepine receptor with a high affinity, should be initiated, thus inhibiting the action of these drugs[48] (Table 3).

Table 3 Treatment strategies in patients with precipitant-induced episodic HE.
General supportive care
Prevention of falls or body harm in disorientated patients
Care of bladder and bowel function
Care of i.v. lines
Monitor fluid balance
Monitor glycaemia and electrolytes
Monitor arterial blood gases
Correct acid/base disturbances
Monitor blood pressure
Avoid aspiration pneumonia
Prevent causes of sepsis
Support nutritional needsAn energy intake of 35-40 kcal /kg BW/d and a protein intake of 1.2-1.5 g/kg BW/d are recommended. Energy should be provided by glucose and fat in a ratio of 65-50: 35%-50% of non protein calories according to the ESPEN guidelines for nutrition in liver disease (31) In patients with severe hepatic encephalopathy (Grade III-IV), solutions with an increase content of BCAAs and reduced amount of aromatic amino acid can ameliorate neurological symptoms ensuring adequate protein intake
Treatment of the precipitating event
GI bleedingStop bleeding with vasoactive drugs, endoscopic therapy or angiographic shunt (TIPS) Correct anaemia with blood transfusion Nasogastric tube to facilitate upper GI cleansing
Infection (pulmonary, urinary tract, spontaneous bacterial peritonitis)Appropriate antibiotic terapie
Exogenous sedativesDiscontinue benzodiazepines
Electrolyte abnormalitiesDiscontinue diuretics Correct hypo or hyperkalemia
ConstipationCathartic Bowel enema
Deterioration of renal functionDiscontinue diuretics Correct dehydration Discontinue nephrotoxic antibiotics
Recurrent/persistent HE

Searching for possible identifiable and correctable precipitating events is important also in recurrent/persistent HE. The presence of large spontaneous porto-systemic shunts is demonstrable in some patients with persistent HE[21]. In selected cases, the diameter of such large shunts have been reduced by surgical or radiological techniques, resulting in an amelioration in HE[49-52]. Since large spontaneous shunts are similar to a well-functioning surgical or radiological anastomosis, this approach must be balanced against the possibility of a significant increase in portal hypertension and liver transplantation should always be considered as the first option in these patients. Refractory post-TIPS HE may also be successfully treated by reducing the stent diameter[22,25]. Since the treatment should be prolonged life-long, the ideal therapy should be extremely safe and well tolerated.

PHARMACOLOGICAL APPROACHES TO THE TREATMENT OF HE

The following pharmacological approaches have been used in the treatment of HE (Table 4).

Table 4 Some of the RCTs available on the treatment of hepatic encephalopathy.
Ref.Type of studyType of HE/patients (n)Tested treatmentControl treatmentResults
Uribe et al[23] 1987Double-blind, controlled trialEpisodic overt HE (20)Lactitol and lactose enemasNonacidifying enemasAcidifying agents like lactose and lactitol are effective and superior to tap-water enemas for the treatment of HE
Hassanein et al[31] 2007Prospective, randomized, controlled, multicenter trialSevere episodic overt HE (70)Molecular adsorbent recirculating system (MARS) and standard medical therapyStandard medical therapyMARS is associated with an earlier and more frequent improvement of HE
Kircheis et al[57] 1997Randomized, double-blind, placebo-controlled, multicenter trialMHE (53) and mild overt HE (grade I-II, 53)L-ornithine-L-aspartate, 20 g/d i.v.PlaceboTherapy improves psychometric performance and is safe and effective in HE treatment
Stauch et al[58] 1998Randomized, double-blind, placebo-controlled clinical trialMHE (23) and mild overt HE (43)L-ornithine-L-aspartate, 9 g/d orallyPlaceboTherapy improves psychometric performance, blood ammonia levels and is safe and effective in HE treatment
Ahmad et al[59] 2008RCTOvert HE (80)L-ornithine-L-aspartate, 20 g/d i.v.PlaceboTreatment improves blood ammonia and mental state
Sushma et al[62] 1992Prospective randomized double-blind studyOvert HE (74)Sodium Benzoate, 5 gLactuloseImprovement in portal-systemic encephalopathy parameters occurred in both treatment groups and was similar
Reding et al[66] 1984Double-blind randomised trialChronic HE (22)Zinc acetate 600 mg/dPlaceboTreatment improves psychometric performance
Riggio et al[67] 1991Double-blind, crossover trialChronic HE (15)Zinc sulfate 600 mg/d and lactitolLactitolZinc was significantly raised after oral administration, but no modification in the parameters included in Conn's index were observed
Loguercio et al[72] 1995Double blind parallel trialHE (40)SF-68LactuloseSF-68 improves blood ammonia and psychometric performance
Gentile[75] 2005Randomized, double-blind, crossover studyHE (107)Acarbose 300 mg/dPlaceboAcarbose improves blood ammonia levels and HE clinical parameters
Low-absorbable antibiotics

One of the first strategies applied in patients with HE aimed to suppress bacteria involved in colonic ammonia genesis. In fact, more than 25 trials evaluated the use of a large series of antibiotics (i.e. neomycin, paromomycin, metronidazole, vancomycin, rifaximin) in patients with minimal, persistent and episodic HE. Currently, however, the use of antibiotics is only recommended for short periods of time. However, a recent study suggested that rifaximine may be given for prolonged periods safely and this drug may be able to prevent HE recurrence in patients with previous episodes of OHE[53].

Non-absorbable disaccharides

Lactulose and lactitol administered per os, reaching the colon unmodified, act by reducing the production and absorption of ammonia through several mechanisms. A recent systematic review concluded that “there is insufficient evidence to determine whether non-absorbable disaccharides are of benefit in patients with HE”[54]. Although this analysis can be criticized on several aspects[55], it has raised the issue of non-absorbable disaccharide efficacy and, more generally, the need for placebo-controlled trials in the therapy of HE[56]. An open label RCT that was recently published concluded that lactulose is effective for secondary prophylaxis of HE; i.e in patients who recovered after a first episode[39].

Interestingly, in the group of patients treated with lactulose, significantly fewer patients developed an overt episode of HE precipitated by infections. Non-absorbable disaccharides were able to reduce bacterial translocation in cirrhotic patients, thus reducing the occurrence of spontaneous bacterial peritonitis and other infections. By the same mechanism, these drugs were able to reduce the inflammation, which had been proposed as a pathogenetic mechanism for astrocyte swelling in the brain.

Association between intestinal antibiotics and non-absorbable disaccharides

From a pathophysiological point of view, the association between intestinal antibiotics and non-absorbable disaccharides should not provide additive benefits because the former are supposed to act against the intestinal flora, which are responsible for the efficacy of the latter. Nonetheless, some clinical observations support a role for this association in the sub-group of patients with a scarce response to each agent separately. The efficacy of the association can be monitored through the measurement of faecal pH, which should be maintained as lower than 6.

Ornithine aspartate

L-ornithine-L-aspartate (LOLA) is a stable salt composed of two natural amino acids, both important substrates in the metabolic conversion of ammonia to urea and glutamine. LOLA induces an increase of liver and muscle ammonia metabolism, leading to a decrease in blood levels, and is able to cross the blood-brain barrier, increasing the cerebral ammonia disposal. The drug can be administrated both intravenously and orally[57-60]. According to the results of published studies, LOLA is more effective than placebo in reducing blood ammonia and in ameliorating patient mental status and psychometric performance. It should be noted that additional studies have been completed but the publication of results was not allowed by the company producing the drug[55]. Moreover, LOLA is not available in several countries. Recently LOLA showed similar results to placebo in patients with acute liver failure and HE[61].

Drugs favoring alternative pathways of nitrogen metabolism

Sodium benzoate (SB) is able to bind ammonia to form hippurate, a non-toxic substance that can be eliminated in urine and is widely used in patients with a congenital deficit in the urea cycle. SB has the advantage of being particularly inexpensive (1/30 of the cost of lactulose). SB showed an efficacy similar to lactulose in patients with episodic precipitant-induced HE[62], but a note of caution in the use of SB in cirrhotic patients derives from the observation that its administration (10 g/d) induced an increase in both basal blood ammonia and glutamine-induced ammonia levels[63]. Moreover, the prolonged use of SB can interfere with the management of ascites because it induces a significant sodium load. Sodium phenylacetate and SB (Ammonul) is the only drug approved by the Food and Drug Administration for the treatment of acute hyperammonemia and associated encephalopathy in patients with urea cycle disorders[64]. Ammonul via intravenous injections is able to promote the synthesis of non-urea nitrogen-containing metabolites, which can be excreted in the urine. A RCT on cirrhotic patients with severe (grade III-IV) HE is currently ongoing. HPN-100 is a pro-drug of phenylbutyrate and a pre-pro-drug of phenylacetic acid (dosed orally in a liquid form). HPN-100 provides an alternative pathway to the urea cycle for the disposal of waste nitrogen through renal excretion of phenylacetylglutamine. The drug is under investigation in patients with urea cycle diseases but has never been used in patients with liver failure.

Zinc

Zinc deficiency has been reported in patients with liver cirrhosis and related neurologic dysfunction. Moreover, reduced zinc concentrations inversely correlate with blood ammonia and experimental studies showed that zinc supplementation improves ammonia detoxification through urea genesis by increasing liver ornithine transcarbamylase activity[65]. Existing RCTs on oral zinc supplementation to cirrhotic patients, however, have shown conflicting results[66,67].

Benzodiazepine antagonists

Endogenous benzodiazepine (BZD)-like substances have been involved in HE pathogenesis. Flumazenil is a competitive BZD antagonist. One meta-analysis of six RCTs[68] showed that flumazenil induced a clinical improvement in 27% of the patients versus 3% of the placebo group, and an improvement of the EEG tracing (19% vs 2%). Another meta-analysis, including 12 trials for a total of 756 patients, led to the conclusion that flumazenil had no significant effect on recovery or survival from HE[69]. No side effects were observed during flumazenil treatment.

BCAAs

Several RCTs and reviews have assessed the effects of BCAAs in patients with HE. In fact, solutions with an increased content of BCAAs and a reduced amount of aromatic amino acids can ameliorate neurological symptoms by ensuring an adequate protein intake at the same time. One meta-analysis[70] concluded that BCAA increased recovery rates from episodic HE without definite effects on mortality, and a more recent review[71], based on 11 RCTs, where BCAAs were either parenterally or orally administered and given alone or as part of solutions containing other amino acids, confirmed that BCAAs significantly increased the number of patients improving from HE as compared with control treatments, without any convincing evidence of any effect on survival. The authors remarked that most trials were small, with a short follow-up, and were of low methodological quality.

Modulators of intestinal bacterial flora

Probiotics, synbiotics (when combined with fermentable fibres), and prebiotics can reduce ammonia production by modifying the gut bacterial flora. The effect of these products in the treatment of HE are currently under investigation: two RCTs have been reported on the effect of probiotics on minimal[41] and OHE[72]; two on synbiotics in MHE[73,74] and one on prebiotic acarbose in patients with OHE[75]. Table 5 shows an overview of the new therapeutic approaches for HE treatment.

Table 5 Possible future approaches to HE treatment.
ObjectivesApproaches currently usedProposed approaches under evaluation
To reduce the production of Gut-derived toxinsDisaccharides Low-absorbable antibioticsProbiotics Fermentable fibers, acarbose Spherical adsorptive carbon (AST 120)
To favor nitrogen metabolismLiver transplantation Reduction of TIPS diameter Closure of a spontaneous portal systemic shunt Ornithine Aspartate Zinc Sodium benzoateArtificial liver support Sodium benzoate + phenylacetate (Ammonul) L-ornithine phenylacetate HPN-100 (Phenylbutyrate + phenylacetic acid)
To correct the alterations in neurotransmissionBCAA Flumazenil
FURTHER APPROACHES
Artificial devices

Artificial devices have been shown to improve HE symptoms in patients with decompensated cirrhosis, probably by favoring the disposal of toxins accumulated as the result of a failing liver. Extra-corporeal albumin dialysis (ECAD), using the molecular adsorbent re-circulating system (MARS), has been recently tested by a RCT. Seventy patients with grade-III or grade-IV HE were randomised to receive ECAD plus standard medical therapy (SMT) or SMT alone. The improvement of HE was significantly more frequent and faster in the ECAD group as compared with the SMT group[31].

Correction of electrolyte disturbances

Hyponatremia, commonly found in patients with cirrhosis and ascites, may increase brain edema, a typical condition in case of acute liver failure. A “low-grade brain edema” leading to astrocyte swelling has been also hypothetically thought to be involved in the pathogenesis of type-C HE[29,76]. In rats with a portacaval shunt, chronic hyponatremia exacerbated ammonia-induced brain edema[77]. Hyponatremia was related, in patients with cirrhosis, to the comparison of electroencephalographic abnormalities, known as a risk factor for the development of OHE[22] and a well-known precipitating factor for the development of episodic HE. In this direction, Aquaretic drugs have been proposed in the treatment of hyponatremia in cirrhotic patients[78]. Unfortunately, their use in the prevention of HE in cirrhotic patients with low sodium levels or in the treatment of episodic HE precipitated by hyponatremia can be only hypothesized, since it was never tested until recently. No data are available on the evolution of HE in patients treated with aquaretic drugs.

CONCLUSION

The efficacy of most pharmacological approaches traditionally used in the treatment of HE is still a matter of intense debate. The empirically developed strategies for the treatment of HE, such as the identification and treatment of the precipitating event and the general support of the patients with episodic HE or the reduction of the blood flow thought a large portal-systemic shunt in the patients with recurrent/persistent HE, make this complication treatable in the majority of patients.

Therapy for HE remains strongly based on the pathophysiological assumption that nitrogen substances coming from the gut are not cleared by the failing liver and are relevant for the modifications occurring in the central nervous system. Improvement in our knowledge of the pathogenesis of HE suggests that several new therapies for HE will emerge (Table 5). These new approaches are likely to be derived from: (1) a better understanding of the cause of astrocyte swelling in the brain as well as the role of inflammation in the pathogenesis of HE; and (2) new knowledge on ammonia trafficking and on the role of intestine and kidney in nitrogen handling. At the same time, our competence in testing old and new treatment modalities should be enhanced urgently. Novel, well designed studies are needed. In particular, RCTs of the new era should have the following characteristics: (1) Homogeneous patients should be included. For example, the inclusion of patient groups with both minimal and OHE in the same study is to be avoided, as it is now clear that these two types of patients are not comparable and the methodology used to stage their symptoms is completely different; (2) Appropriate end points for the study need to be chosen, such as improvement in quality of life or the prevention of future OHE manifestations; and (3) It is necessary to standardize and objectively stage the severity of OHE. In fact, the development of a simple and clinically applicable standardized grading scale, useful for both diagnosing and staging, is essential to obtain a diagnostic tool that is easily applied in practice and sufficiently accurate to offer precise end-points for controlled therapeutic trials.

Footnotes

Peer reviewers: George Papatheodoridis, Assistant Professor, Department of Internal Medicine, Hippokration Hospital of Athens, 114 Vas. Sofia save., 11527 Athens, Greece; Spilios Manolakopoulos, Professor, Department of Medicine, Hippokration General Hospital, 3 Vironos street, 15343, Agia Paraskevi, Athens, Greece

S- Editor Li LF L- Editor Lutze M E- Editor Yang C

References
1.  Ferenci P, Lockwood A, Mullen K, Tarter R, Weissenborn K, Blei AT. Hepatic encephalopathy--definition, nomenclature, diagnosis, and quantification: final report of the working party at the 11th World Congresses of Gastroenterology, Vienna, 1998. Hepatology. 2002;35:716-721.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Amodio P, Montagnese S, Gatta A, Morgan MY. Characteristics of minimal hepatic encephalopathy. Metab Brain Dis. 2004;19:253-267.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Quero JC, Hartmann IJ, Meulstee J, Hop WC, Schalm SW. The diagnosis of subclinical hepatic encephalopathy in patients with cirrhosis using neuropsychological tests and automated electroencephalogram analysis. Hepatology. 1996;24:556-560.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Amodio P, Del Piccolo F, Marchetti P, Angeli P, Iemmolo R, Caregaro L, Merkel C, Gerunda G, Gatta A. Clinical features and survivial of cirrhotic patients with subclinical cognitive alterations detected by the number connection test and computerized psychometric tests. Hepatology. 1999;29:1662-1667.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Groeneweg M, Moerland W, Quero JC, Hop WC, Krabbe PF, Schalm SW. Screening of subclinical hepatic encephalopathy. J Hepatol. 2000;32:748-753.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Amodio P, Del Piccolo F, Pettenò E, Mapelli D, Angeli P, Iemmolo R, Muraca M, Musto C, Gerunda G, Rizzo C. Prevalence and prognostic value of quantified electroencephalogram (EEG) alterations in cirrhotic patients. J Hepatol. 2001;35:37-45.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Bajaj JS, Wade JB, Sanyal AJ. Spectrum of neurocognitive impairment in cirrhosis: Implications for the assessment of hepatic encephalopathy. Hepatology. 2009;50:2014-2021.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Ripoll C, Groszmann R, Garcia-Tsao G, Grace N, Burroughs A, Planas R, Escorsell A, Garcia-Pagan JC, Makuch R, Patch D. Hepatic venous pressure gradient predicts clinical decompensation in patients with compensated cirrhosis. Gastroenterology. 2007;133:481-488.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Sanyal AJ, Freedman AM, Shiffman ML, Purdum PP 3rd, Luketic VA, Cheatham AK. Portosystemic encephalopathy after transjugular intrahepatic portosystemic shunt: results of a prospective controlled study. Hepatology. 1994;20:46-55.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Rössle M, Haag K, Ochs A, Sellinger M, Nöldge G, Perarnau JM, Berger E, Blum U, Gabelmann A, Hauenstein K. The transjugular intrahepatic portosystemic stent-shunt procedure for variceal bleeding. N Engl J Med. 1994;330:165-171.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Riggio O, Merlli M, Pedretti G, Servi R, Meddi P, Lionetti R, Rossi P, Bezzi M, Salvatori F, Ugolotti U. Hepatic encephalopathy after transjugular intrahepatic portosystemic shunt. Incidence and risk factors. Dig Dis Sci. 1996;41:578-584.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Salerno F, Borroni G, Moser P, Badalamenti S, Cassarà L, Maggi A, Fusini M, Cesana B. Survival and prognostic factors of cirrhotic patients with ascites: a study of 134 outpatients. Am J Gastroenterol. 1993;88:514-519.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Merkel C, Bolognesi M, Angeli P, Noventa F, Caregaro L, Sacerdoti D, Gatta A. Prognostic indicators of survival in patients with cirrhosis and esophageal varices, without previous bleeding. Am J Gastroenterol. 1989;84:717-722.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Christensen E, Krintel JJ, Hansen SM, Johansen JK, Juhl E. Prognosis after the first episode of gastrointestinal bleeding or coma in cirrhosis. Survival and prognostic factors. Scand J Gastroenterol. 1989;24:999-1006.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Bajaj JS. Minimal hepatic encephalopathy matters in daily life. World J Gastroenterol. 2008;14:3609-3615.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Bajaj JS, Hafeezullah M, Hoffmann RG, Varma RR, Franco J, Binion DG, Hammeke TA, Saeian K. Navigation skill impairment: Another dimension of the driving difficulties in minimal hepatic encephalopathy. Hepatology. 2008;47:596-604.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Bajaj JS, Saeian K, Schubert CM, Hafeezullah M, Franco J, Varma RR, Gibson DP, Hoffmann RG, Stravitz RT, Heuman DM. Minimal hepatic encephalopathy is associated with motor vehicle crashes: the reality beyond the driving test. Hepatology. 2009;50:1175-1183.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Kircheis G, Knoche A, Hilger N, Manhart F, Schnitzler A, Schulze H, Häussinger D. Hepatic encephalopathy and fitness to drive. Gastroenterology. 2009;137:1706-1715.e1-9.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Klempnaue J, Schrem H. Review: surgical shunts and encephalopathy. Metab Brain Dis. 2001;16:21-25.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Somberg KA, Riegler JL, LaBerge JM, Doherty-Simor MM, Bachetti P, Roberts JP, Lake JR. Hepatic encephalopathy after transjugular intrahepatic portosystemic shunts: incidence and risk factors. Am J Gastroenterol. 1995;90:549-555.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Riggio O, Efrati C, Catalano C, Pediconi F, Mecarelli O, Accornero N, Nicolao F, Angeloni S, Masini A, Ridola L. High prevalence of spontaneous portal-systemic shunts in persistent hepatic encephalopathy: a case-control study. Hepatology. 2005;42:1158-1165.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Riggio O, Angeloni S, Salvatori FM, De Santis A, Cerini F, Farcomeni A, Attili AF, Merli M. Incidence, natural history, and risk factors of hepatic encephalopathy after transjugular intrahepatic portosystemic shunt with polytetrafluoroethylene-covered stent grafts. Am J Gastroenterol. 2008;103:2738-2746.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Uribe M, Campollo O, Vargas F, Ravelli GP, Mundo F, Zapata L, Gil S, Garcia-Ramos G. Acidifying enemas (lactitol and lactose) vs. nonacidifying enemas (tap water) to treat acute portal-systemic encephalopathy: a double-blind, randomized clinical trial. Hepatology. 1987;7:639-643.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Rolachon A, Zarski JP, Lutz JM, Fournet J, Hostein J. [Is the intestinal lavage with a solution of mannitol effective in the prevention of post-hemorrhagic hepatic encephalopathy in patients with liver cirrhosis? Results of a randomized prospective study]. Gastroenterol Clin Biol. 1994;18:1057-1062.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Riggio O, Nicolao F, Angeloni S, Masini A, Salvatori F, Fanelli F, Efrati C, Merli M. Intractable hepatic encephalopathy after tips with polytetrafluoroethylene-covered stent-graft. Scand J Gastroenterol. 2003;38:570-572.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Uflacker R, d'Albuquerque LA, de Oliveira e Silva A, de Freitas JM, Gama-Rodrigues JJ. Embolization to reverse severe recurrent hepatic encephalopathy. Arq Gastroenterol. 1988;25 Spec No:21-25.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Schliess F, Görg B, Häussinger D. RNA oxidation and zinc in hepatic encephalopathy and hyperammonemia. Metab Brain Dis. 2009;24:119-134.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Norenberg MD, Jayakumar AR, Rama Rao KV, Panickar KS. New concepts in the mechanism of ammonia-induced astrocyte swelling. Metab Brain Dis. 2007;22:219-234.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Häussinger D, Schliess F. Pathogenetic mechanisms of hepatic encephalopathy. Gut. 2008;57:1156-1165.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Shawcross DL, Shabbir SS, Taylor NJ, Hughes RD. Ammonia and the neutrophil in the pathogenesis of hepatic encephalopathy in cirrhosis. Hepatology. 2010;51:1062-1069.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Hassanein TI, Tofteng F, Brown RS Jr, McGuire B, Lynch P, Mehta R, Larsen FS, Gornbein J, Stange J, Blei AT. Randomized controlled study of extracorporeal albumin dialysis for hepatic encephalopathy in advanced cirrhosis. Hepatology. 2007;46:1853-1862.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Ghanta RK, Mullen KD. Placebo controlled treatment trials in hepatic encephalopathy are feasible and ethical. Hepatology. 2005;42:1206-1207.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Merli M, Riggio O, Dally L. Does malnutrition affect survival in cirrhosis? PINC (Policentrica Italiana Nutrizione Cirrosi). Hepatology. 1996;23:1041-1046.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Rodríguez D, Martín A, Martinez-Valle F. Prognosis after surviving to the first episode of hepatic encephalopathy [Abstract]. Hepatology. 1998;28:268.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Hartmann IJ, Groeneweg M, Quero JC, Beijeman SJ, de Man RA, Hop WC, Schalm SW. The prognostic significance of subclinical hepatic encephalopathy. Am J Gastroenterol. 2000;95:2029-2034.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Romero-Gómez M, Boza F, García-Valdecasas MS, García E, Aguilar-Reina J. Subclinical hepatic encephalopathy predicts the development of overt hepatic encephalopathy. Am J Gastroenterol. 2001;96:2718-2723.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Blei AT, Córdoba J. Hepatic Encephalopathy. Am J Gastroenterol. 2001;96:1968-1976.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Riggio O, Masini A, Efrati C, Nicolao F, Angeloni S, Salvatori FM, Bezzi M, Attili AF, Merli M. Pharmacological prophylaxis of hepatic encephalopathy after transjugular intrahepatic portosystemic shunt: a randomized controlled study. J Hepatol. 2005;42:674-679.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Sharma BC, Sharma P, Agrawal A, Sarin SK. Secondary prophylaxis of hepatic encephalopathy: an open-label randomized controlled trial of lactulose versus placebo. Gastroenterology. 2009;137:885-891, 891.e1.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Romero-Gomez M, Jover M, Diaz-Gomez D, de Teran LC, Rodrigo R, Camacho I, Echevarria M, Felipo V, Bautista JD. Phosphate-activated glutaminase activity is enhanced in brain, intestine and kidneys of rats following portacaval anastomosis. World J Gastroenterol. 2006;12:2406-2411.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Bajaj JS, Saeian K, Christensen KM, Hafeezullah M, Varma RR, Franco J, Pleuss JA, Krakower G, Hoffmann RG, Binion DG. Probiotic yogurt for the treatment of minimal hepatic encephalopathy. Am J Gastroenterol. 2008;103:1707-1015.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Kanematsu T, Koyanagi N, Matsumata T, Kitano S, Takenaka K, Sugimachi K. Lack of preventive effect of branched-chain amino acid solution on postoperative hepatic encephalopathy in patients with cirrhosis: a randomized, prospective trial. Surgery. 1988;104:482-488.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Bajaj JS, Etemadian A, Hafeezullah M, Saeian K. Testing for minimal hepatic encephalopathy in the United States: An AASLD survey. Hepatology. 2007;45:833-834.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Bajaj JS, Hafeezullah M, Franco J, Varma RR, Hoffmann RG, Knox JF, Hischke D, Hammeke TA, Pinkerton SD, Saeian K. Inhibitory control test for the diagnosis of minimal hepatic encephalopathy. Gastroenterology. 2008;135:1591-1600.e1.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Mardini H, Saxby BK, Record CO. Computerized psychometric testing in minimal encephalopathy and modulation by nitrogen challenge and liver transplant. Gastroenterology. 2008;135:1582-1590.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Prasad S, Dhiman RK, Duseja A, Chawla YK, Sharma A, Agarwal R. Lactulose improves cognitive functions and health-related quality of life in patients with cirrhosis who have minimal hepatic encephalopathy. Hepatology. 2007;45:549-559.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Plauth M, Cabré E, Riggio O, Assis-Camilo M, Pirlich M, Kondrup J, Ferenci P, Holm E, Vom Dahl S, Müller MJ. ESPEN Guidelines on Enteral Nutrition: Liver disease. Clin Nutr. 2006;25:285-294.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Barbaro G, Di Lorenzo G, Soldini M, Giancaspro G, Bellomo G, Belloni G, Grisorio B, Annese M, Bacca D, Francavilla R. Flumazenil for hepatic encephalopathy grade III and IVa in patients with cirrhosis: an Italian multicenter double-blind, placebo-controlled, cross-over study. Hepatology. 1998;28:374-378.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Sakurabayashi S, Sezai S, Yamamoto Y, Hirano M, Oka H. Embolization of portal-systemic shunts in cirrhotic patients with chronic recurrent hepatic encephalopathy. Cardiovasc Intervent Radiol. 1997;20:120-124.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Kato T, Uematsu T, Nishigaki Y, Sugihara J, Tomita E, Moriwaki H. Therapeutic effect of balloon-occluded retrograde transvenous obliteration on portal-systemic encephalopathy in patients with liver cirrhosis. Intern Med. 2001;40:688-691.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Uflacker R, Silva Ade O, d'Albuquerque LA, Piske RL, Mourão GS. Chronic portosystemic encephalopathy: embolization of portosystemic shunts. Radiology. 1987;165:721-725.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Córdoba J, Olivé G, Alonso J, Rovira A, Segarra A, Pérez M, Jacas C, Vargas V. Improvement of magnetic resonance spectroscopic abnormalities but not pallidal hyperintensity followed amelioration of hepatic encephalopathy after occlusion of a large spleno-renal shunt. J Hepatol. 2001;34:176-178.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Bass NM, Mullen KD, Sanyal A, Poordad F, Neff G, Leevy CB, Sigal S, Sheikh MY, Beavers K, Frederick T. Rifaximin treatment in hepatic encephalopathy. N Engl J Med. 2010;362:1071-1081.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Als-Nielsen B, Gluud LL, Gluud C. Non-absorbable disaccharides for hepatic encephalopathy: systematic review of randomised trials. BMJ. 2004;328:1046.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Morgan MY, Blei A, Grüngreiff K, Jalan R, Kircheis G, Marchesini G, Riggio O, Weissenborn K. The treatment of hepatic encephalopathy. Metab Brain Dis. 2007;22:389-405.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Shawcross D, Jalan R. Dispelling myths in the treatment of hepatic encephalopathy. Lancet. 2005;365:431-433.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Kircheis G, Nilius R, Held C, Berndt H, Buchner M, Görtelmeyer R, Hendricks R, Krüger B, Kuklinski B, Meister H. Therapeutic efficacy of L-ornithine-L-aspartate infusions in patients with cirrhosis and hepatic encephalopathy: results of a placebo-controlled, double-blind study. Hepatology. 1997;25:1351-1360.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Stauch S, Kircheis G, Adler G, Beckh K, Ditschuneit H, Görtelmeyer R, Hendricks R, Heuser A, Karoff C, Malfertheiner P. Oral L-ornithine-L-aspartate therapy of chronic hepatic encephalopathy: results of a placebo-controlled double-blind study. J Hepatol. 1998;28:856-864.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Ahmad I, Khan AA, Alam A, Dilshad A, Butt AK, Shafqat F, Malik K, Sarwar S. L-ornithine-L-aspartate infusion efficacy in hepatic encephalopathy. J Coll Physicians Surg Pak. 2008;18:684-687.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Poo JL, Góngora J, Sánchez-Avila F, Aguilar-Castillo S, García-Ramos G, Fernández-Zertuche M, Rodríguez-Fragoso L, Uribe M. Efficacy of oral L-ornithine-L-aspartate in cirrhotic patients with hyperammonemic hepatic encephalopathy. Results of a randomized, lactulose-controlled study. Ann Hepatol. 2006;5:281-288.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Acharya SK, Bhatia V, Sreenivas V, Khanal S, Panda SK. Efficacy of L-ornithine L-aspartate in acute liver failure: a double-blind, randomized, placebo-controlled study. Gastroenterology. 2009;136:2159-2168.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Sushma S, Dasarathy S, Tandon RK, Jain S, Gupta S, Bhist MS. Sodium benzoate in the treatment of acute hepatic encephalopathy: a double-blind randomized trial. Hepatology. 1992;16:138-144.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Efrati C, Masini A, Merli M, Valeriano V, Riggio O. Effect of sodium benzoate on blood ammonia response to oral glutamine challenge in cirrhotic patients: a note of caution. Am J Gastroenterol. 2000;95:3574-3578.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Jalan R, Wright G, Davies NA, Hodges SJ. L-Ornithine phenylacetate (OP): a novel treatment for hyperammonemia and hepatic encephalopathy. Med Hypotheses. 2007;69:1064-1069.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Riggio O, Merli M, Capocaccia L, Caschera M, Zullo A, Pinto G, Gaudio E, Franchitto A, Spagnoli R, D'Aquilino E. Zinc supplementation reduces blood ammonia and increases liver ornithine transcarbamylase activity in experimental cirrhosis. Hepatology. 1992;16:785-789.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Reding P, Duchateau J, Bataille C. Oral zinc supplementation improves hepatic encephalopathy. Results of a randomised controlled trial. Lancet. 1984;2:493-495.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Riggio O, Ariosto F, Merli M, Caschera M, Zullo A, Balducci G, Ziparo V, Pedretti G, Fiaccadori F, Bottari E. Short-term oral zinc supplementation does not improve chronic hepatic encephalopathy. Results of a double-blind crossover trial. Dig Dis Sci. 1991;36:1204-1208.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Goulenok C, Bernard B, Cadranel JF, Thabut D, Di Martino V, Opolon P, Poynard T. Flumazenil vs. placebo in hepatic encephalopathy in patients with cirrhosis: a meta-analysis. Aliment Pharmacol Ther. 2002;16:361-372.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Als-Nielsen B, Kjaergard LL, Gluud C. Benzodiazepine receptor antagonists for acute and chronic hepatic encephalopathy. Cochrane Database Syst Rev. 2001;CD002798.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Naylor CD, O'Rourke K, Detsky AS, Baker JP. Parenteral nutrition with branched-chain amino acids in hepatic encephalopathy. A meta-analysis. Gastroenterology. 1989;97:1033-1042.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Als-Nielsen B, Koretz RL, Kjaergard LL, Gluud C. Branched-chain amino acids for hepatic encephalopathy. Cochrane Database Syst Rev. 2003;CD001939.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Loguercio C, Abbiati R, Rinaldi M, Romano A, Del Vecchio Blanco C, Coltorti M. Long-term effects of Enterococcus faecium SF68 versus lactulose in the treatment of patients with cirrhosis and grade 1-2 hepatic encephalopathy. J Hepatol. 1995;23:39-46.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Liu Q, Duan ZP, Ha DK, Bengmark S, Kurtovic J, Riordan SM. Synbiotic modulation of gut flora: effect on minimal hepatic encephalopathy in patients with cirrhosis. Hepatology. 2004;39:1441-1449.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Malaguarnera M, Gargante MP, Malaguarnera G, Salmeri M, Mastrojeni S, Rampello L, Pennisi G, Li Volti G, Galvano F. Bifidobacterium combined with fructo-oligosaccharide versus lactulose in the treatment of patients with hepatic encephalopathy. Eur J Gastroenterol Hepatol. 2010;22:199-206.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Gentile S, Guarino G, Romano M, Alagia IA, Fierro M, Annunziata S, Magliano PL, Gravina AG, Torella R. A randomized controlled trial of acarbose in hepatic encephalopathy. Clin Gastroenterol Hepatol. 2005;3:184-191.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Guevara M, Baccaro ME, Torre A, Gómez-Ansón B, Ríos J, Torres F, Rami L, Monté-Rubio GC, Martín-Llahí M, Arroyo V. Hyponatremia is a risk factor of hepatic encephalopathy in patients with cirrhosis: a prospective study with time-dependent analysis. Am J Gastroenterol. 2009;104:1382-1389.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Córdoba J, Gottstein J, Blei AT. Chronic hyponatremia exacerbates ammonia-induced brain edema in rats after portacaval anastomosis. J Hepatol. 1998;29:589-594.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Ginès P, Guevara M. Hyponatremia in cirrhosis: pathogenesis, clinical significance, and management. Hepatology. 2008;48:1002-1010.  [PubMed]  [DOI]  [Cited in This Article: ]