[N A J Med Sci. 2016;9(2):59-65. DOI: 10.7156/najms.2016.0902059]

Zhixian Guo; Zujiang Yu; Keqin Hu, MD

Hepatic encephalopathy (HE) is one of the most serious complications of chronic or fulminant liver failure. It’s main clinical manifestations involving disturbance of consciousness, behavioral disorders and coma. According to the degree of impaired consciousness, neurological signs and EEG changes, the clinical course of HE may be divided into four phases, ranging from cognitive alteration to coma and even death, staging contribute to early diagnosis, prognosis and therapeutic evaluation. Management of HE primarily involves avoidance of precipitating factors, protect live function from further damage, treatment of ammonia poisoning and regulation of neurotransmitter. This review mainly discusses the current available options for therapy in Hepatic encephalopathy and some new studied agents.

Key Words: hepatic encephalopathy, treatment, nonabsorbable disaccharides, antibiotics

INTRODUCTION Hepatic encephalopathy (HE) is a reversible and devastating complication of advanced liver disease, occurs in approximately 30-45% of patients with cirrhosis.1-3 Compared with cirrhotic patients who have not had HE, the patients who had HE accounts for frequent hospitalization, decrease in the quality of life, and poorer outcomes. The high morbidity and mortality combined with the costs underline the importance of effective treatment and prevention of HE.

The clinical manifestations of HE range from altered mental status to deep coma and even death,4 the West Haven scale are most often used to grade HE, the scale ranges from trivial lack of awareness, alteration in sleep-wake cycle to coma.5 Current treatment strategies are aimed at reducing the serum level of ammonia. This is done by introducing agents that reduce or inhibit production of intestinal ammonia or minimize its absorption from the gastrointestinal tract and correcting precipitating factors such as gastrointestinal hemorrhage, electrolyte imbalances and constipation.

CLASSIFICATION OF HE According to the Working Party at the 11th World Congress of Gastroenterology, HE is classified into three different types.4 Type A describes HE associated with acute liver failure. Type B describes HE associated with portal-systemic shunting without associated intrinsic liver disease. Type C describes HE associated with underlying cirrhosis and portal hypertension or portal-systemic shunts. Type C is further subcategorized as covert HE (minimal HE and West-Haven grade I HE) and overt HE (West-Haven grades II-IV HE).

CHE can affect up to 80% of cirrhotic patients. Because of lack of clinical symptoms, it is difficult to diagnosis. It can only be diagnosed through specialized neurophysiologic and psychometric tests.6 Patients with CHE have defects in attention, vigilance and orientation. It is very dangerous for them to drive.7 It negatively affects the patients’ quality of life and their families. CHE is associated with mortality and increased risk of hospitalization and OHE development.8,9 We will discuss CHE later. Here, we mainly discuss OHE.

PATHOGENESIS OF HE The pathogenesis of HE is multifactorial, and is not completely understood, but the elevated blood ammonia levels are a key factor in the process of the disease.10 Ammonia is mainly generated in the intestine as a result of bacterial breakdown of dietary protein metabolism of glutamine.11 The absorption of ammonia occurs in the colon via nonionic diffusion and is transported through the portal vein to the liver.12 Hyperammonemia induces encephalopathy by promoting cerebral edema, modulating the blood-brain barrier, decreasing blood flow, or modulating neuroinhibition. Recently, more and more people demonstrated that inflammation,13,14 including systemic inflammation, neuroinflammation, oxidative stress,15 and neurosteroids,16 acts in concert with ammonia in the pathogenesis of HE. But they are not well understood by now. Therefore, a number of treatments aim at lowering the level of blood ammonia.17

MANAGEMENT OF HE The management of HE is mainly supportive care, primarily involves identification and treatment of underlying etiologies and precipitating factors, and administration of various ammonia-lowering therapies.

Effectively Treaing Underlying Etiologies for Hepatic Decompensation or Liver Failure Although most unserlying eitologies are difficult to manage, especially in the acute care setting, some of these etiologies could be treated simultaneously. For instance, HBV and HCV cirrhosis is no reversiable; acetaminophen overdose should be treated immediately on admission. Alcohol liver disease could be another sample that can be treated. Final para of this session: for those liver injury is chronic and cirrhosis is advanced, especially those with high MELD score, liver Tx evaluation and listing should also be considered.

Early Identification and Prompt Treatment of Precipitating Factors The main step in treatment of HE is identifying and treating precipitating factors.18 Commonly precipitating factors are listed in table 1. It is estimated that over 80% of HE patients are caused by reversible factors.19 In clinical, Infection is very common in HE patients, all patients with ascites should have a diagnostic paracentesis, if infection is found, it should be treated with appropriate antibiotics. Constipation is also very easy to be found, it is important to assess recent bowel habit, it is appropriate to produce two to three soft bowel movements per day. Blood biochemical test results can indicate electrolyte abnormality. Addressing these factors in time have been proved to be crucial in effectively treating most patients with HE.20

Table 1. Precipitating factors for hepatic encephalopathy
Drugs benzodiazepines narcotics alcohol Hypovolemia diureses, diarrhea vomit, bleeding large volume paracentesis
Increase the production and absorption of ammonia excessive dietary protein gastrointestinal bleeding infections constipation hypokalemia Portosystemic shunting surgery or natural diversion
Angiemphraxis portal thrombosis hepatic vein thrombosis
Hepatocellular carcinoma

Reduce the Generation and Absorption of Nitrogen in the Intestine Diet Management. Diet management is very important in the management of HE.21 Usually, protein-restricted diets are prescribed for cirrhotic patients with hepatic encephalopathy.22 However, protein restriction may worsen the patient’s nutritional status.23 A randomized study has proved that the outcome of hepatic encephalopathy was not significantly different between low-protein diet group and normal-protein diet group, they also proved that protein synthesis was similar for low and normal protein diet, but those of the low-protein diet group showed higher protein breakdown.24 Clinical guidelines recommend protein intake begins at a dose of 0.5g/kg/day, with progressive increase to 1-1.5g/kg/day.25 Vegetable protein may be better than animal protein.26 In addition, high fiber diet may be helpful in decreasing the colonic transit time and absorption of ammonia. Small meals evenly distributed throughout the day and a late-night snack of complex carbohydrates will help minimize protein utilization.27

Nonabsorbable Disaccharides. Nonabsorbable disaccharides are the most accepted and widely used treatments for HE.28,29 Nonabsorbable disaccharides, such as lactulose and lactitol, are synthetic disaccharides, they cannot be broken down in the small intestine after oral administration, after reaching the colon, they can be decomposed into lactic acid and acetic acid to reduce the pH value of the intestinal tract.30 Acidified colon encourage the conversion of ammonia to ammonium, which is more membrane impermeable, as a result, less ammonia is absorbed by the colon.31 Moreover, the acidification of gut lumen makes the enteric environment less suitable for ammoniagenic coliform bacteria and leads to increased levels of non-ammoniagenic lactobacilli.19

The side effect of lactulose therapy include flatulence, stomachache, nausea, emesis and so on.32 Besides, its taste sweet and greasy, makes it can’t be accepted by every patient. The optimal dose of lactulose is that the patient has two or three soft stools each day.33 It can also be used to retention enema. The curative effect of lactitol is similar with lactulose, but its low sweetness, good taste and less adverse reaction, makes it more acceptable than lactulose.34

The curative effect of nonabsorbable disaccharides is exactly, they can be used for the treatment of every stage of hepatic encephalopathy and minimal hepatic encephalopathy.35 A 2004 meta-analysis reported that nonabsorbable disaccharides were superior to placebo but did not improve survival. However, not all patients respond to lactulose, Praveen Sharma and his workmates analysed 231 cirrhosis patients with HE, they all treated with lactulose and correction of any associated precipitating factors, they concluded that high baseline MELD, high total leukocyte count, low serum sodium, low MAP, and presence of hepatocellular carcinoma were predictors of nonresponse to lactulose.36 Besides Nielsen et al found that non-absorbable disaccharides were inferior to antibiotics in reducing the risk of no improvement and lowering blood ammonia concentration, they concluded that there was insufficient evidence to support or refute the use of non-absorbable disaccharides for hepatic encephalopathy.37 Lactulose or lactitol is recommended by clinical guidelines as the first line therapy, anyway.28

Antibiotics. The antibiotic therapies can reduce the enteric bacterial flora that may play a vital role in the production of neurotoxins leading to encephalopathy.38,39 Commonly used antibiotics are neomycin, metronidazole, rifaximin and so on. Even though neomycin and metronidazole have been shown to be as effective as lactulose,40 their side effect profile, such as ototoxicity and nephrotoxicity (neomycin) and neurotoxicity (metronidazole), limits their use, particularly in the long term.41 Rifaximin is a synthetic antimicrobial, it is poorly absorbed in gastrointestinal tract, it has a broad spectrum of antibacterial action covering Gram-positive and Gram-negative organisms, both aerobes and anaerobes.42 It binds to the b-subunit of the bacterial DNA dependent RNA polymerase and disrupts RNA synthesis. The therapeutic effect of rifaximin is similar with neomycin.43,44 But the side effect of rifaximin is minimal, for example, headache, flatulence, abdominal pain, constipation, nausea, and vomiting, and no reported drug interactions make it relatively safe.45,46

Over recent years, rifaximin has become the most widely used antibiotic in the empiric and prophylactic treatment of HE due to its limited side effect profile. And many studies have demonstrated the efficacy of rifaximin in the management of hepatic encephalopathy (Table 2).

Table 2. The data.
Trial Study design Assessment Cases Duration of treatment Conclusions
Loguercio et al 200348 Prospective, randomized Cancellation test, Reitan test, EEG, and PSE severity 40 3 months Rifaximin>lactulose
Paik et al 200587 Prospective, randomized Gradation of blood ammonia, flapping tremor, mental status, number connection test 54 7 days Rifaximin=lactulose
Jiang et al 200888 Retrospective, randomized, comparative - 264 - Rifaximin=nonabsorbable disaccharides
Bass et al 201089 Prospective, randomized Conn score and asterixis grade 299 6 months Rifaximin>placebo
Sanyal et al 201190 Prospective, randomized Conn score and asterixis grade 219 6 months Rifaximin>placebo
Eltawil et al 201291 Retrospective, randomized, comparative Serum ammonia levels, mental status, asterixis 565 - Rifaximin=disaccharides
Mullen et al 201492 Prospective, randomized Adverse events, clinical laboratory parameters 392 6 months Rifaximin>placebo
Bajaj et al 201593 Prospective, randomized Rates of HE events, rates of HE-related hospitalisation 82 6 months Rifaximin>placebo

Combination Therapy. Lactulose and rifaximin have shown to be effective in HE, to evaluate the efficacy and safety of lactulose plus rifiximin vs. lactulose alone in the management of HE, Sharma et al conducted a prospective double-blind randomized controlled trial, in this trial, 120 patients were randomized into two groups, group A (lactulose plus rifiximin) and group B (lactulose plus placebo). They found that there was a significant decrease in mortality after treatment with lactulose plus rifaximin vs. lactulose and placebo, and there were more deaths in group B, however, there were no differences in gastrointestinal bleed and hepatorenal syndrome, but it also proved that combination of lactulose plus rifaximin is effective than lactulose alone.47 To evaluate the effect of rifaximin, lactitol and their combination in treating chronic HE, Loguercio et al randomly assigned forty out-patients in different groups, HE was assessed by considering: mental state, asterixis, number connection test, arterial blood ammonia levels. They concluded that rifaximin in combination with lactitol or sorbitol represents an effective and safe treatment of chronic HE.48

In a retrospective study, Mohammad et al conducted a PubMed MEDLINE search, the key words included lactulose, lactitol, nonabsorbable disaccharide, metronidazole, rifaximin, neomycin, probiotics, and hepatic encephalopathy. They found 6 studies and concluded that the combination of rifaximin and lactulose should be considered in the treatment and prevention of HE.49 The unfortunate reality is that there are no available clinical studies evaluating dual antibiotic therapy, nonabsorbable disaccharides with probiotics, or antibiotics with probitics.

Probiotics. The effectiveness of lactulose and rifaximin suggest that altering gut flora to a non-urease-producing population may be effective in managing HE.50 Probiotics alter the intestinal microbiota and reduce the production of ammonia.51 It has been shown that the administration of probiotics to liver cirrhotic patients resulted in a modulation of the gut flora with a significant reduction of the quantity of several bacterial pathogens in fecal bacteriological analysis.52

To assess the effect of probiotic therapies on HE in liver cirrhotic patients, Jun Xu et al collected six randomized controlled trials involving 496 liver cirrhotic patients. The results showed that probiotic therapy significantly reduced the development of overt hepatic encephalopathy (OR [95% CI]: 0.42 [0.26, 0.70], P = 0.0007). However, probiotics did not affect mortality, levels of serum ammonia and constipation (mortality: OR [95% CI]: 0.73 [0.38, 1.41], P = 0.35; serum ammonia: WMD [95% CI]: -3.67 [-15.71, 8.37], P = 0.55; constipation: OR [95% CI]: 0.67 [0.29, 1.56], P = 0.35). 53 While another randomized trial come to the similar conclusions.54 Before probitics can be endorsed as effective therapy for hepatic encephalopathy, demonstration of unequivocal efficacy is needed.

L-Ornithine L-aspartate. L-Ornithine L-aspartate(LOLA), ornithine and aspartate are substrates of the urea cycle.55 Even in decompensated cirrhosis, it seems to stimulate the enzyme activity in residual hepatocytes.56 LOLA decreases ammonia levels by stimulating hepatic urea cycle activity and promoting glutamine synthesis.57

Is reported in many studies, both oral and intravenous forms of LOLA have the efficacy to decrease ammonia levels and improve HE.58,59 A meta-analysis of eight randomized controlled trials with 646 patients showed that, compared with placebo/no-intervention control, LOLA benefits both overt and minimal HE patients, and significantly reduced fasting ammonia levels. To critically evaluate the efficacy of the use of LOLA when compared to placebo in the treatment of HE, four studies published between 1993 and 2000 were selected and reviewed, the trials proved the efficacy of LOLA in reducing hyperammonemia of hepatic encephalopathy.60 A meta-analysis of 217 patients proved the efficacy of LOLA in reducing hyperammonemia of HE again, but they also found that there is no sufficient evidence of a significant beneficial effect of LOLA on patients with HE.60 So there is a lack of consensus related to the usage of LOLA in HE now.61

Branch Chain Amino Acids. Branch Chain Amino Acid(BCAA) preparation is a compound amino acids, it is mainly composed of leucine, isoleucine and valine.62 The mechanism of BCAA is competitive inhibit the aromatic amino acids to enter in the brain, and reduce the formation of false neurotransmitters.63,64 The curative effect of BCAA is still controversial.65 But for malnutrition who intolerance to protein, supply BCAA helps to improve the negative nitrogen balance.66 Besides, BCAA supplementation may improve albumin synthesis, decrease insulin resistance, decrease hepatocellular carcinoma, and improve immune function.67 Supplementation can be done via oral or intravenous routes. A double-blind placebo-controlled crossover study indicates that long-term branched-chain amino acid supplementation is well tolerated and effective in the treatment of impaired automobile driving capacity associated with latent HE.68 While Als-Nielsen et al analyzed eleven randomized trials which involving 556 patients, they found no convincing evidence that BCAA had a significant beneficial effect on patients with hepatic encephalopathy.69 These contradiction may underlie the difficulty in assessing the clinical effects of BCAAs.

Molecular Adsorbent Recirculating System. Molecular adsorbent recirculating system (MARS) is a blood detoxification system that removes fat-soluble, water-soluble and albumin-bound toxins by means of albumin recycling system, activated carbon, resin and dialysis.70,71 This system can remove a part of toxic substances in the blood of patients with hepatic encephalopathy, decrease the concentration of serum bilirubin and improve the prothrombin time, and have a temporary and a certain degree of curative effect to hepatic encephalopathy, at the same time it may be possible to win the time for liver transplantation.72

To prove the efficacy, safety, and tolerability of MARS, Hassanein et al conducted a prospective, randomized, controlled, multicenter trial in severe hepatic encephalopathy patients. Patients were randomized to MARS and standard medical therapy (SMT) or SMT alone, the results indicated that the improvement proportion of HE was higher in MARS versus the SMT group and was reached faster and more frequently than in the SMT group, besides, the subjects receiving MARS tolerated treatment well with no unexpected adverse events.73 Exclusion criteria in this trial included active hemorrhage, active infections, severe cardiopulmonary disease and so on, which will decrease the applicability of MARS.74 The insufficient of this study was not designed to examine the impact of MARS on survival, a full assessment of the role of albumin dialysis awaits the results of additional controlled trials.75

Zinc. Zinc is a critical cofactor in the metabolism of ammonia, zinc deficiency is associated with the down-regulation of muscle glutamine synthetase, which leads to hyperammonemia.76 A study aim to assess serum zinc levels in a cohort of healthy subjects and cirrhotic patients, they found that in cirrhotic patients zinc serum levels were significantly lower than in healthy subjects, and a stepwise decline in serum zinc with worsening Child class.77 Reding et al conducted a double-blind randomized trial in which 22 cirrhotic patients with HE were given oral zinc supplementation or placebo. In the group which received zinc acetate 600mg/day for 7 days, serum zinc had been restored to normal by day 8. On day 8, HE was improved in the supplemented group but not in the placebo group. Besides, blood urea nitrogen was also significant increase in the supplemented group.78,79 But the duration of this improvement requires further investigation.80-82 A meta-analysis of 233 patients revealed that oral zinc supplementation improved performance on the number connection test, but there was no evidence about other clinical or biochemical outcomes.83 Therefore more trials are needed to evaluate the efficacy of oral zinc acetate in the patients with liver cirrhosis and hepatic encephalopathy.

Sodium Benzoate. Sodium benzoate is thought to activate a non-urea cycle pathway for ammonia removal.84 To evaluate the efficacy of sodium benzoate in the management of HE, Sushma et al conducted a prospective randomized double-blind study, the patients were randomized to receive sodium benzoate (5 gm twice daily) or lactulose (dose adjusted for 2 or 3 semiformed stools/day), the results showed that sodium benzoate is a safe and effective alternative to lactulose in the treatment of acute HE.85 Given the need for intravenous administration, and the dose-dependent sodium content in this therapy, it may not be useful in treating HE.86

SUMMARY Hepatic encephalopathy (HE) is one of the most serious complications of chronic or fulminant liver failure, and encompasses a spectrum of neuropsychiatric symptoms and signs. Due to differences in etiology and severity, as well as heterogeneity of manifestations, the diagnosis and management of HE remain a difficult challenge for physicians and medical professionals. Several factors have been implicated in pathogenesis, with ammonia considered to have a central role, thus prompt reduce the level of ammonia can be potentially lifesaving. Identification and correction of precipitating factors remains the cornerstone of treatment. Lactulose and rifaximin are the two most effective therapeutic agents available now, and we believe that lactulose plus rifaximin will obtain a better treatment effect. While in clinical practice, to reduce morbidity and mortality, we also combination with other agents, probiotics, L-Ornithine L-aspartate, Branch Chain Amino Acid and so on, although there are no enough evidences to prove the effectiveness of these drugs. It is clear that little progress has been made in developing new therapeutic options over the last 20 years, so study the mechanism of HE and find new drugs are very important now. Overall, when we meet in clinical patients with hepatic encephalopathy, prompt correction of precipitating factors, decrease blood and cerebral ammonia levels quickly, use antibiotics properly and apply molecular adsorbent recirculating system when necessary. If these are not work, liver transplant may be a choice.

___________________________________________________________________________

Zhixian Guo;1 Zujiang Yu;1* Keqin Hu, MD2* 1Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China 2Division of Gastroenterology and Hepatology Univ. of California, Irvine Medical Center, CA *Corresponding Author: Division of Gastroenterology and Hepatology Univ. of California, Irvine Medical Center, CA. (Email: kqhu@uci.edu, johnyuem@zzu.edu.cn) CONFLICT OF INTEREST The authors have no conflict of interest to disclose. ___________________________________________________________________________ REFERENCES
  1. Poordad FF. Review article: the burden of hepatic encephalopathy. Aliment Pharmacol Ther. 2007;25(Suppl 1):3-9.
  2. D'Amico G, Garcia-Tsao G, Pagliaro L. Natural history and prognostic indicators of survival in cirrhosis: a systematic review of 118 studies. J Hepatol. 2006;44:217-231.
  3. Eroglu Y, Byrne WJ. Hepatic encephalopathy. Emerg Med Clin North Am. 2009;27:401-414.
  4. Ferenci P. Hepatic encephalopathy--definition, nomenclature, diagnosis, and quantification: final report of the working party at the 11th World Congresses of Gastroenterology, Vienna, 1998. Hepatology (Baltimore, Md.) 35, 716-721 (2002).
  5. Vilstrup H. Hepatic encephalopathy in chronic liver disease: 2014 Practice Guideline by the American Association for the Study of Liver Diseases and the European Association for the Study of the Liver. Hepatology. 2014;60:715-735.
  6. Yen CL, Liaw YF. Somatosensory evoked potentials and number connection test in the detection of subclinical hepatic encephalopathy. Hepatogastroenterology. 1990;37:332-334.
  7. Schomerus H. Latent portasystemic encephalopathy. I. Nature of cerebral functional defects and their effect on fitness to drive. Dig Dis Sci. 1981;26:622-630.
  8. Patidar KR. Covert hepatic encephalopathy is independently associated with poor survival and increased risk of hospitalization. Am J Gastroenterol. 2014;109:1757-1763.
  9. Hartmann IJ. The prognostic significance of subclinical hepatic encephalopathy. Am J Gastroenterol. 2000;95:2029-2034.
  10. Butterworth RF, Giguere JF, Michaud J, Lavoie J, Layrargues GP. Ammonia: key factor in the pathogenesis of hepatic encephalopathy. Neurochem Pathol. 1987;6:1-12.
  11. Windmueller HG, Spaeth AE. Uptake and metabolism of plasma glutamine by the small intestine. J Biol Chem. 1974;249:5070-5079.
  12. Castell DO, Moore EW. Ammonia absorption from the human colon. The role of nonionic diffusion. Gastroenterology. 1971;60:33-42.
  13. Luo M, Guo JY, Cao WK. Inflammation: A novel target of current therapies for hepatic encephalopathy in liver cirrhosis. World J Gastroenterol. 2015;21:11815-11824.
  14. Milewski K, Oria M. What we know: the inflammatory basis of hepatic encephalopathy. Metab Brain Dis. 2015. [Epub ahead of print]
  15. Bemeur C, Desjardins P, Butterworth RF. Evidence for oxidative/nitrosative stress in the pathogenesis of hepatic encephalopathy. Metab Brain Dis. 2010;25:3-9.
  16. Butterworth RF. Neurosteroids in hepatic encephalopathy: Novel insights and new therapeutic opportunities. J Steroid Biochem Mol Biol. 2015. [Epub ahead of print]
  17. Petersen KU. Options in the treatment of hepatic encephalopathy. Medizinische Monatsschrift fur Pharmazeuten. 38, 160-164 (2015).
  18. Hepatic encephalopathy. Early therapy is most important!. MMW Fortschritte der Medizin. 2014;156:28.
  19. Toris GT, Bikis CN, Tsourouflis GS, Theocharis SE. Hepatic encephalopathy: an updated approach from pathogenesis to treatment. Med Sci Monit. 2011;17:Ra53-63.
  20. Bresci G, Parisi G, Banti S. Management of hepatic encephalopathy with oral zinc supplementation: a long-term treatment. Eur J Med. 1993;2:414-416.
  21. Amodio P, Canesso F, Montagnese S. Dietary management of hepatic encephalopathy revisited. Curr Opin Clin Nutr Metab Care. 2014;17:448-452.
  22. Yang HC, Lin SY. Evidence-based clinical decision making: dietary protein intake recommendations for hepatic encephalopathy patients. The Journal of Nursing. 2013;60:90-96.
  23. Jurado Garcia J, Costan Rodero G, Calanas-Continente A. Importance of nutritional support in patients with hepatic encephalopathy. Nutr Hosp. 2012;27:372-381.
  24. Cordoba J. Normal protein diet for episodic hepatic encephalopathy: results of a randomized study. J Hepatol. 2004;41:38-43.
  25. Antar R, Wong P, Ghali P. A meta-analysis of nutritional supplementation for management of hospitalized alcoholic hepatitis. Can J Gastroenterol. 2012;26:463-467.
  26. Bianchi GP. Vegetable versus animal protein diet in cirrhotic patients with chronic encephalopathy. A randomized cross-over comparison. J Int Med. 1993;233:385-392.
  27. Amodio P. The nutritional management of hepatic encephalopathy in patients with cirrhosis: International Society for Hepatic Encephalopathy and Nitrogen Metabolism Consensus. Hepatology. 2013;58:325-336.
  28. Blei AT, Cordoba J. Hepatic Encephalopathy. Am J Gastroenterol. 2001;96:1968-1976.
  29. Riordan, S.M. & Williams, R. Treatment of hepatic encephalopathy. The New England journal of medicine 337, 473-479 (1997).
  30. Sharma, P. & Sharma, B.C. Disaccharides in the treatment of hepatic encephalopathy. Metabolic brain disease 28, 313-320 (2013).
  31. Rahimi RS, Singal AG, Cuthbert JA, Rockey DC. Lactulose vs polyethylene glycol 3350--electrolyte solution for treatment of overt hepatic encephalopathy: the HELP randomized clinical trial. JAMA. 2014;174:1727-1733.
  32. Guslandi M, Cella A. Rifaximin and nonabsorbable disaccharides for hepatic encephalopathy. Eur J Gastroenterol Hepatol. 2010;22:376.
  33. Als-Nielsen BE, Gluud LL, Gluud CN. Nonabsorbable disaccharides for the treatment of hepatic encephalopathy--a systemic review of randomized clinical trials--a secondary publication. Ugeskr Laeger. 2005;167:179-182.
  34. Morgan MY, Hawley KE. Lactitol vs. lactulose in the treatment of acute hepatic encephalopathy in cirrhotic patients: a double-blind, randomized trial. Hepatology. 1987;7:1278-1284.
  35. Als-Nielsen B, Gluud LL, Gluud C. Nonabsorbable disaccharides for hepatic encephalopathy. Cochrane Database Syst Rev. 2004; (2):CD003044.
  36. Sharma P, Sharma BC, Sarin SK. Predictors of nonresponse to lactulose in patients with cirrhosis and hepatic encephalopathy. Eur J Gastroenterol Hepatol. 2010;22:526-531.
  37. Als-Nielsen B, Gluud LL, Gluud C. Non-absorbable disaccharides for hepatic encephalopathy: systematic review of randomised trials. BMJ. 2004;328:1046.
  38. DuPont HL. Rifaximin: An Antibiotic with Important Biologic Effects. Mini Rev Med Chem. 2015;16:200-205.
  39. Hirota SA. Understanding the molecular mechanisms of rifaximin in the treatment of gastrointestinal disorders - a focus on the modulation of host tissue function. Mini Rev Med Chem. 2015;16:206-217.
  40. Conn HO. Comparison of lactulose and neomycin in the treatment of chronic portal-systemic encephalopathy. A double blind controlled trial. Gastroenterology. 1977;72:573-583.
  41. Leise MD, Poterucha JJ, Kamath PS, Kim WR. Management of hepatic encephalopathy in the hospital. Mayo Clin Proc. 2014;89:241-253.
  42. Sharma P, Sharma BC. Management of overt hepatic encephalopathy. J Clin Exp Hepatol. 2015;5:S82-87.
  43. Pedretti G, Calzetti C, Missale G, Fiaccadori F. Rifaximin versus neomycin on hyperammoniemia in chronic portal systemic encephalopathy of cirrhotics. A double-blind, randomized trial. Ital J Gastroenterol. 1991;23:175-178.
  44. Di Piazza, S. et al. Rifaximine versus neomycin in the treatment of portosystemic encephalopathy. Ital J Gastroenterol. 1991;23:403-407.
  45. Sanchez-Delgado J, Miquel M. Role of rifaximin in the treatment of hepatic encephalopathy. Gastroenterol Hepatol. 2016;39:282-292.
  46. Wahib AA. Evaluation of rifaximin in management of hepatic encephalopathy. J Egypt Soc Parasitol. 2014;44:677-685.
  47. Sharma BC. A randomized, double-blind, controlled trial comparing rifaximin plus lactulose with lactulose alone in treatment of overt hepatic encephalopathy. The American journal of gastroenterology 108, 1458-1463 (2013).
  48. Loguercio C, Federico A, De Girolamo V, Ferrieri A, Del Vecchio Blanco C. Cyclic treatment of chronic hepatic encephalopathy with rifaximin. Results of a double-blind clinical study. Minerva gastroenterologica e dietologica 49, 53-62 (2003).
  49. Mohammad RA, Regal RE, Alaniz C. Combination therapy for the treatment and prevention of hepatic encephalopathy. The Annals of pharmacotherapy 46, 1559-1563 (2012).
  50. Saab S. Probiotics are Helpful in Hepatic Encephalopathy: A Meta-Analysis of Randomized Trials. Liver international : official journal of the International Association for the Study of the Liver (2015).
  51. Solga, S.F. Probiotics can treat hepatic encephalopathy. Medical hypotheses 61, 307-313 (2003).
  52. Liu Q. Synbiotic modulation of gut flora: effect on minimal hepatic encephalopathy in patients with cirrhosis. Hepatology (Baltimore, Md.) 39, 1441-1449 (2004).
  53. Xu J. Effects of probiotic therapy on hepatic encephalopathy in patients with liver cirrhosis: an updated meta-analysis of six randomized controlled trials. Hepatobiliary & pancreatic diseases international: HBPD INT. 13, 354-360 (2014).
  54. McGee RG, Bakens A, Wiley K, Riordan SM, Webster AC. Probiotics for patients with hepatic encephalopathy. The Cochrane database of systematic reviews, Cd008716 (2011).
  55. Blanco Vela, Poo Ramirez JL. Efficacy of oral L-ornithine L-aspartate in cirrhotic patients with hyperammonemic hepatic encephalopathy. Annals of hepatology 10 Suppl 2, S55-59 (2011).
  56. Zhou ZW. Ornithine aspartate and naloxone combined therapy for hepatic encephalopathy affects cognitive function, prognosis, and neuropeptide levels. Zhonghua gan zang bing za zhi = Zhonghua ganzangbing zazhi = Chinese journal of hepatology 21, 385-388 (2013).
  57. Rose C. L-ornithine-L-aspartate in experimental portal-systemic encephalopathy: therapeutic efficacy and mechanism of action. Metabolic brain disease 13, 147-157 (1998).
  58. Kircheis Gl. Therapeutic efficacy of L-ornithine-L-aspartate infusions in patients with cirrhosis and hepatic encephalopathy: results of a placebo-controlled, double-blind study. Hepatology (Baltimore, Md.) 25, 1351-1360 (1997).
  59. Stauch S. Oral L-ornithine-L-aspartate therapy of chronic hepatic encephalopathy: results of a placebo-controlled double-blind study. J Hepatol. 1998;28:856-864.
  60. Soarez PC, Oliveira AC, Padovan J, Parise ER, Ferraz MB. A critical analysis of studies assessing L-ornithine-L-aspartate (LOLA) in hepatic encephalopathy treatment. Arquivos de Gastroenterologia 46, 241-247 (2009).
  61. Bai M. Randomised clinical trial: L-ornithine-L-aspartate reduces significantly the increase of venous ammonia concentration after TIPSS. Aliment Pharmacol Ther. 2014;40:63-71.
  62. Gluud LL. Oral branched-chain amino acids have a beneficial effect on manifestations of hepatic encephalopathy in a systematic review with meta-analyses of randomized controlled trials. J Nutri. 2013;143:1263-1268.
  63. Holecek M. Three targets of branched-chain amino acid supplementation in the treatment of liver disease. Nutrition. 2010;26:482-490.
  64. Gluud, L.L. et al. Branched-chain amino acids for people with hepatic encephalopathy. Cochrane Database Syst Rev. 2015;9:CD001939.
  65. Kawaguchi T, Taniguchi E, Sata M. Effects of oral branched-chain amino acids on hepatic encephalopathy and outcome in patients with liver cirrhosis. Nutr Clin Pract. 2013;28:580-588.
  66. Ndraha S, Hasan I, Simadibrata M. The effect of L-ornithine L-aspartate and branch chain amino acids on encephalopathy and nutritional status in liver cirrhosis with malnutrition. Acta Med Indones. 2011;43:18-22.
  67. Kawaguchi T, Izumi N, Charlton MR, Sata M. Branched-chain amino acids as pharmacological nutrients in chronic liver disease. Hepatology. 2011;54:1063-1070.
  68. Plauth M. Long-term treatment of latent portosystemic encephalopathy with branched-chain amino acids. A double-blind placebo-controlled crossover study. J Hepatol. 1993;17:308-314.
  69. Als-Nielsen B, Koretz RL, Kjaergard LL, Gluud C. Branched-chain amino acids for hepatic encephalopathy. Cochrane Database Syst Rev; 2003:CD001939.
  70. Banares R, Catalina MV, Vaquero J. Molecular adsorbent recirculating system and bioartificial devices for liver failure. Clin Liver Dis. 2014;18:945-956.
  71. Kobashi-Margain RA. Albumin dialysis with molecular adsorbent recirculating system (MARS) for the treatment of hepatic encephalopathy in liver failure. Ann Hepatol. 2011;10(Suppl 2):S70-76.
  72. Steiner C, Mitzner S. Experiences with MARS liver support therapy in liver failure: analysis of 176 patients of the International MARS Registry. Liver. 2002;22(Suppl 2):20-25.
  73. Hassanein TI. Randomized controlled study of extracorporeal albumin dialysis for hepatic encephalopathy in advanced cirrhosis. Hepatology. 2007;46:1853-1862.
  74. Leise MD. Treatment of persistent/medically refractory covert hepatic encephalopathy with the molecular adsorbent recirculating system. Liver Transpl. 2014;20:867-868.
  75. Cisneros-Garza LE. The molecular adsorbent recirculating system as a liver support system: summary of Mexican experience. Ann Hepatol. 2014;13:240-247.
  76. Schliess F, Gorg B, Haussinger D. RNA oxidation and zinc in hepatic encephalopathy and hyperammonemia. Metab Brain Dis. 2009;24:119-134.
  77. Poo JL. Serum zinc concentrations in two cohorts of 153 healthy subjects and 100 cirrhotic patients from Mexico City. Digestive diseases. 2015;3:136-142.
  78. Reding P, Duchateau J, Bataille C. Oral zinc supplementation improves hepatic encephalopathy. Results of a randomised controlled trial. Lancet. 1984;2:493-495.
  79. Hayashi M, Ikezawa K, Ono A, et al. Evaluation of the effects of combination therapy with branched-chain amino acid and zinc supplements on nitrogen metabolism in liver cirrhosis. Hepatol Res. 2007;37:615-619.
  80. Tuerk MJ Fazel N. Zinc deficiency. Curr Opin Gastroenterol. 2009;25:136-143.
  81. Marchesini G, Fabbri A, Bianchi G, Brizi M, Zoli M. Zinc supplementation and amino acid-nitrogen metabolism in patients with advanced cirrhosis. Hepatology. 1996;23:1084-1092.
  82. Takeda A. Zinc homeostasis and functions of zinc in the brain. Biometals. 2001;14:343-351.
  83. Chavez-Tapia NC. A systematic review and meta-analysis of the use of oral zinc in the treatment of hepatic encephalopathy. Nutr J. 2013;12:74.
  84. Misel ML, Gish RG, Patton H, Mendler, M. Sodium benzoate for treatment of hepatic encephalopathy. Gastroenterol Hepatol. 2013;9:219-227.
  85. Sushma S. Sodium benzoate in the treatment of acute hepatic encephalopathy: a double-blind randomized trial. Hepatology. 1992;16:138-144.
  86. Matoori S, Leroux JC. Recent advances in the treatment of hyperammonemia. Adv Drug Deliv Rev. 2015;90:55-68.
  87. Paik YH. Comparison of rifaximin and lactulose for the treatment of hepatic encephalopathy: a prospective randomized study. Yonsei Med J. 2005;46:399-407.
  88. Jiang Q. Rifaximin versus nonabsorbable disaccharides in the management of hepatic encephalopathy: a meta-analysis. Eur J Gastroenterol Hepatol. 2008;20:1064-1070.
  89. Bass NM. Rifaximin treatment in hepatic encephalopathy. N Engl J Med. 2010;362:1071-1081.
  90. Sanyal A. Randomised clinical trial: rifaximin improves health-related quality of life in cirrhotic patients with hepatic encephalopathy - a double-blind placebo-controlled study. Aliment Pharmacol Ther. 2011;34:853-861.
  91. Eltawil, K.M., Laryea, M., Peltekian, K. & Molinari, M. Rifaximin vs. conventional oral therapy for hepatic encephalopathy: a meta-analysis. World J Gastroenterol. 2012;18:767-777.
  92. Mullen, K.D. et al. Rifaximin is safe and well tolerated for long-term maintenance of remission from overt hepatic encephalopathy. Clin Gastroenterol Hepatol. 2014;12:1390-1397.
  93. Bajaj JS, Barrett AC, Bortey E, Paterson C, Forbes WP. Prolonged remission from hepatic encephalopathy with rifaximin: results of a placebo crossover analysis. Aliment Pharmacol Ther. 2015;41:39-45.

Refbacks

  • There are currently no refbacks.