July 24, 2010

HCV Raises Risk of Stroke, Maybe MI, in HIV-Positive US Veterans

Author: Mark Mascolini

12 July 2010

In US veterans with HIV, hepatitis C virus (HCV) coinfection independently raised the risk of cerebrovascular disease (such as stroke) and marginally upped the risk of acute myocardial infarction (MI). Because the veterans population is largely male and has free access to care, it is unclear how closely the findings apply to other HIV-positive populations.

HCV is associated with lower cholesterol levels, but how HCV infection affects risk of MI and cerebrovascular disease in people with HIV had not been well studied. This analysis involved 19,424 HIV-positive veterans, 6183 of them (31.6%) with HCV infection. The study focused on veterans cared for during the earlier part of the current antiretroviral era: 1996 to 2004.

Compared with veterans infected only with HIV, those also infected with HCV were significantly less likely to have high cholesterol (18.0% versus 30.7%, P < 0.001). But HCV-coinfected veterans were more likely to have hypertension (43.8% versus 35.6%, P < 0.001), more likely to have type 2 diabetes mellitus (16.2% versus 11.1%, P < 0.0001), and more likely to smoke (36.7% versus 24.7%, P = 0.009).

In analyses not adjusted for other risk factors, rates of acute MI and cerebrovascular disease were significantly higher in HCV-coinfected veterans than in veterans without HCV: 4.19 versus 3.36 events per 1000 patient-years for acute MI (P < 0.001) and 12.47 versus 11.2 events per 1000 patient-years for cerebrovascular disease (P < 0.001).

A statistical analysis that factored in diabetes, hypertension, age, and duration of antiretroviral therapy determined that cerebrovascular disease was 20% more likely in veterans with HCV than in those without HCV (hazard ratio [HR] 1.20, 95% confidence interval [CI] 1.04 to 1.38), and this increased risk was statistically significant (P = 0.013). In this analysis acute MI was 25% more likely in veterans with HCV, but this higher risk fell short of statistical significance (HR 1.25, 95% CI 0.98 to 1.61, P = 0.072).

Acute MI was significantly more likely in veterans with hypertension (HR 2.05, P < 0.001), older age (HR 1.79, P < 0.001), and longer duration of antiretroviral therapy (HR 1.12, P = 0.0411).

Source: R. Bedimo, A.O. Westfall, M. Mugavero, H. Drechsler, N. Khanna, M. Saag. Hepatitis C virus coinfection and the risk of cardiovascular disease among HIV-infected patients. HIV Medicine. 2010; 11: 462-468.

For the study abstract

(Downloading the complete article requires a subscription to HIV Medicine or an online payment; the abstract is free.)

Source

SciClone Announces Enrollment Of First Patient In Its Phase 2 Trial Of SCV-07 In Hepatitis C

Posted by: admin in Pharmacy Drugs on July 18th, 2010

SciClone Pharmaceuticals, Inc. (NASDAQ: SCLN) today announced the enrollment of its first patient at the Atlanta Gastroenterology Associates in Atlanta, GA, in a phase 2 trial of SCV-07 for the treatment of hepatitis C (also known as HCV). This multicenter, multidose, open-label study is designed to evaluate the safety and immunomodulatory effects of SCV-07 as a monotherapy or in combination with ribavirin in non-cirrhotic patients with genotype 1 chronic HCV who have relapsed after at least 44 weeks of treatment with pegylated interferon and ribavirin.

“During our previous phase 2A clinical trial of SCV-07 as a monotherapy to treat patients with chronic hepatitis C infections, we were pleased by the safety data and were very encouraged by the efficacy signal, namely, a reduction of viral loads and a corresponding increase in neopterin concentration in some patients after only seven days of SCV-07 administration,” said Friedhelm Blobel, Ph.D., President and Chief Executive Officer of SciClone. “SciClone is eager to investigate further SCV-07’s potential to enhance the immune response against hepatitis C and to determine whether the compound is capable of improving the current standard of care treatment.”

“Currently approved therapies for the treatment of HCV can have significant side effects and often fall short of providing the most important treatment outcome, sustained viral response,” said Kenneth Sherman, MD, PhD, Gould Professor of Medicine, Department of Internal Medicine at the University of Cincinnati, and a principal investigator in SciClone’s study. “We are very excited by what appears to be SCV-07’s ability to enhance patients’ immune function without adding significant toxicity. Should ongoing clinical trials show the benefits of adding SCV-07 to ribavirin, it has the potential to become incorporated into standard treatment practices in the future.”

The study, which will monitor biomarkers of immune activation and HCV viral load dynamics, will include two treatment cohorts of 20 patients each, who will receive SCV-07 at a dose of either 0.1 mg/kg or 1.0 mg/kg. The treatment period will be approximately eight weeks long, including four weeks of SCV-07 monotherapy followed by four weeks of SCV-07 in combination with ribavirin. In addition, there will be three follow-up visits within seven weeks after the completion of treatment.

For more information on SciClone’s phase 2 trial of SCV-07 in the treatment of HCV, please visit http://www.clinicaltrials.gov/.

About SCV-07

SCV-07 is a small molecule which stimulates the immune system through inhibition of STAT3-dependant signaling and the resulting effects on T-helper 1 cells, which are essential for clearance of viral infections. SCV-07 has shown a good safety profile in several early stage clinical trials in healthy volunteers and subjects with HCV at various doses. SciClone is also carrying out a phase-2, randomized, double-blinded, placebo-controlled trial of SCV-07 for the treatment of oral mucositis in patients with head and neck cancers undergoing chemo-radiation. The topline results from this trial are expected to be announced in the first half of 2010.

SCV-07 is protected by composition of matter patents as well as multiple method of treatment patents. SciClone has exclusive worldwide rights to SCV-07 outside of Russia, where the molecule has recently been approved for stimulation of depressed immune systems.

About the Hepatitis C Virus

HCV is a blood-borne viral disease which causes inflammation of the liver. The World Health Organization estimates that 170 million people worldwide are infected with HCV, and the Centers for Disease Control estimates that approximately 8 to 10 million people are infected with HCV throughout the U.S. and Europe. Of these patients, approximately 85% are chronically infected, and the persistent liver inflammation in chronically infected patients can develop serious complications including cirrhosis of the liver, liver failure, and hepatocellular carcinoma. Only about half of all naive patients treated with current therapy achieve a sustained viral response, and SciClone estimates nearly 1 million HCV patients in the United States alone have failed or will fail current therapy. The market for HCV therapeutics in the three major economic regions of the United States, Europe and Japan is estimated to total approximately $3 billion currently and is expected to grow to approximately $10 billion by 2014.

Source
SciClone Pharmaceuticals

Source

Taking care of your liver

Sunday July 25, 2010

By Dr S.Y. CHONG

The liver is one of the body’s most important organs and needs to be protected at all times.

THE liver is the largest internal organ of the body. This vital organ is crucial to the smooth functioning of the human body.

It metabolises most of the nutrients that are absorbed by the intestine and detoxifies the blood by removing medications, alcohol and potentially harmful chemicals from the bloodstream – processing them chemically so that they can be expelled from the body by the digestive or urinary systems.

The liver also produces clotting factors and other proteins, stores certain vitamins, minerals (including iron) and sugars, regulates fat stores, and controls the production and excretion of cholesterol.

The liver is an amazing organ that can regenerate its cells within a few weeks. In fact, the liver can tolerate a fair amount of “abuse” and will only show signs of injury when the damage is very advanced.

Examples of liver disorders include hepatitis and cirrhosis. Hepatitis is inflammation of the liver and cirrhosis is scarring of the liver. These two conditions may progress to liver cancer if they are not monitored or treated properly.

As the saying goes, prevention is better than cure. Hence, here are some measures you can take to preserve liver health:

Eat right

Poor nutrition rarely causes liver disease but good nutrition in the form of a balanced diet will enable the liver to perform its many various functions efficiently, resulting in better overall health. It can also help liver cells damaged by hepatitis viruses to regenerate, forming new liver cells.

However, whilst it is important to take vitamins and minerals, please note that an excess of Vitamin A is toxic to the liver and should be taken in moderation.

Limit intake of calories

Excess calories in the form of carbohydrates can add to liver dysfunction and can cause fat deposits in the liver, contributing to fatty liver.

No more than 30% of a person’s total calories should come from fat because of the danger to the cardiovascular system. In order to estimate your daily calorie needs, you will need a minimum of 15 calories a day for each pound you weigh.

Watch the alcohol

Liquor, beer and wine are difficult for the liver to metabolise. The daily recommended alcohol intake is three units/drinks for men and two units/drinks for women.

As a general guide, one unit of alcohol translates to half a pint of ordinary strength beer, a small measure (25ml) of spirits or a standard measure (50ml) of fortified wine such as sherry or port.

Having three drinks or more per day should be avoided, as it may lead to alcoholic hepatitis and cirrhosis. People with liver disease should never drink alcohol at all. The same goes for individuals who are taking medication – mixing alcohol with painkillers or other types of medications can be dangerous to your liver.

In particular, the mixture of alcohol and acetaminophen (an ingredient in pain killers and cough medication) can cause sudden, severe hepatitis and even fatal liver failure. If you are not sure which medications to take in combination, please consult your doctor.

Beware ‘nutritional therapies’

Herbal treatments and alternative liver medicines should undergo rigorous scientific study before they can be recommended. “Natural” or diet treatments and herbal remedies can be quite dangerous. Plants of the Crotalaria, Senecio and Heliotopium families, as well as chaparral, mistletoe, skullcap, germander, comfrey, margosa oil, mate tea, Gordolobo yerba tea, pennyroyal, and Jin Blu Huan are all toxic to the liver.

Several scientific studies suggest that substances in milk thistle may protect the liver from harmful substances such as acetaminophen, which can cause liver damage. It is also believed that milk thistle has antioxidant and anti-inflammatory properties, and it may help the liver repair itself by growing new cells.

Stop smoking and stay away from toxic fumes and liquids

Fumes from paint thinners, bug sprays, and other aerosol sprays are picked up by the tiny blood vessels in your lungs and carried to your liver where they are detoxified and discharged in your bile.

The amount and concentration of those chemicals should be controlled to prevent liver damage. Make certain you have good ventilation, use a mask, cover your skin, and wash off any chemicals you get on your skin with soap and water as soon as possible.

Wash your hands

Hands should be washed with soap and water following bowel movements and before food preparation and consumption. This will help prevent the spread of hepatitis A.

Practise safe sex

Wear protection when having intercourse. Hepatitis B and C are transmitted through blood and body fluids. So use condoms and avoid sharing your personal items such as toothbrush, razor or manicure sets, especially if your partner is suffering from a liver disease.

Get vaccinated

Vaccination for hepatitis A and B is available. An immunisation programme for hepatitis B has been in place for all children and adults since 1989 to prevent hepatitis B infection. It is essential to vaccinate newborns for hepatitis B as infections in this group will result in 90% chronic infection. There is no vaccination available for hepatitis C.

In Malaysia, hepatitis B is the most common chronic liver infection, affecting over 1.1 million people. Whilst the majority of people who have acute hepatitis B will overcome the infection, the virus can linger in about 10% of patients for up to six months. Such patients are known as hepatitis B carriers or have chronic hepatitis B infection, depending on levels of virus in the blood and liver enzyme levels.

Chronic liver infection can lead to cirrhosis, where there are areas of scarring and liver cell regeneration within the liver. Cirrhosis can lead to liver cancer, which is often diagnosed too late as few symptoms appear until it has reached an advanced stage. Signs and symptoms of liver cancer include right upper abdominal pain, jaundice (yellowing of the skin), abdominal swelling, weight loss, fatigue, easy bruising or bleeding.

Apart from surgery, for which most liver cancer patients are ineligible due to the advanced stages of the disease at time of diagnosis, there is currently an oral treatment that has been found to be effective in targeting the liver cancer cells specifically and can be used in patients who are unsuitable for surgery.

Sorafenib is the only oral treatment available in Malaysia that is indicated for the treatment of advanced liver cancer. Currently there is a patient assistance programme for sorafenib, which provides drug assistance to patients who meet the eligibility criteria. More information on this programme can be obtained from the Malaysian Liver Foundation, tel: 03-78426101.

Keeping the liver healthy is essential to keeping your entire being healthy, so take good care of your liver, so that it can take care of you.

This article is contributed by Dr S.Y. Chong, medical advisor with Bayer Schering Pharma. This information is provided for educational purposes only and should not be taken in place of a consultation with your doctor. Bayer Schering Pharma disclaims any and all liability for injury or other damages that could result from use of the information obtained from this article.

Source

Changing face of hepatic encephalopathy: Role of inflammation and oxidative stress

ISSN 1007-9327 CN 14-1219/R World J Gastroenterol 2010 July 21; 16(27): 3347-3357

EDITORIAL

Amit S Seyan, Robin D Hughes, Debbie L Shawcross

Amit S Seyan, Robin D Hughes, Debbie L Shawcross, Institute of Liver Studies, King’s College London School of Medicine at King’s College Hospital, King’s College Hospital, Denmark Hill, London, SE5 9RS, United Kingdom

Author contributions: Seyan AS wrote the article; Hughes RD and Shawcross DL edited the article.
Supported by A 5 year UK Department of Health HEFCE Clinical Senior Lectureship (to Dr. Shawcross DL)

Correspondence to: Dr. Debbie L Shawcross, Institute of Liver Studies, King’s College London School of Medicine at King’s College Hospital, King’s College Hospital, Denmark Hill, London, SE5 9RS, United Kingdom. debbie.shawcross@kcl.ac.uk
Telephone: +44-20-32993216
Fax: +44-20-32993167

Received: March 13, 2010
Revised: April 28, 2010
Accepted: May 5, 2010
Published online: July 21, 2010

Abstract

The face of hepatic encephalopathy (HE) is changing. This review explores how this neurocognitive disorder, which is associated with both acute and chronic liver injury, has grown to become a dynamic syndrome that spans a spectrum of neuropsychological impairment, from normal performance to coma. The central role of ammonia in the pathogenesis of HE remains incontrovertible. However, over the past 10 years, the HE community has begun to characterise the key roles of inflammation, infection, and oxidative/nitrosative stress in modulating the pathophysiological effects of ammonia on the astrocyte. This review explores the current thoughts and evidence base in this area and discusses the potential role of existing and novel therapies that might abrogate the oxidative and nitrosative stresses inflicted on the brain in patients with, or at risk of developing, HE.

© 2010 Baishideng. All rights reserved.

Key words: Hepatic encephalopathy; Ammonia; Inflam­mation; Oxidative stress; Astrocyte

Peer reviewers: Dr. Vicente Felipo, Laboratory of Neurobiology, Fundación C.V. Centro de Investigacion Principe Felipe, Avda Autopista del Saler, 16, 46013 Valencia, Spain; Weekitt Kittisupamongkol, MD, Hua Chiew Hospital, Bangkok 10100, Thailand; Xiang-Dong Wang, MD, PhD, Professor, Zhongshan Hospital, Fudan University, Shanghai 230003, China; Dr. Ashraf Dahaba, MD, PhD, MSc, Professor, Department of Anaesthesiology, Medical University of Graz Medical University, Auenbruggerplatz 29, A-8036, Graz, Austria

Seyan AS, Hughes RD, Shawcross DL. Changing face of hepatic encephalopathy: Role of inflammation and oxidative stress. World J Gastroenterol 2010; 16(27): 3347-3357 Available from: URL: http://www.wjgnet.com/1007-9327/full/v16/i27/3347.htm DOI: http://dx.doi.org/10.3748/wjg.v16.i27.3347

INTRODUCTION

Hepatic encephalopathy (HE) is a neurocognitive disorder in which brain function is impaired and is associated with both acute and chronic liver dysfunction. HE occurs in the presence of liver injury or when the liver is bypassed in the presence of a portosystemic shunt. In acute liver failure, patients may develop cerebral oedema and increased intracranial pressure. However, recent studies suggest that intracranial hypertension is less frequent than previously described, complicating 25% of acute cases and only 9% of those with sub-acute liver failure[1]. In cirrhosis, it causes a range of neuropsychiatric and motor disturbances spanning a spectrum of abnormalities, which encompass short-term memory impairment, slowing of reaction time, poor concentration, psychomotor retardation, and sensory dysfunction, through to more clinically apparent neurological signs and symptoms. In its most severe form, patients can develop confusion, stupor, and coma[2]. However, abnormalities can be subtle and only become apparent on formal psychometric testing (minimal HE). Minimal HE is thought to be a disorder of executive functioning, primarily leading to impairments in selective attention, response inhibition, and working memory. This frequently impacts on quality of life[3] and specifically impairs navigation skills[4], which can be demonstrated utilising a driving simulator that correlates impairment with response inhibition and attention[5]. HE has generally been considered to be a reversible process following liver transplantation, although recent studies have suggested that this may not always be the case[6].

The “World Congress of Gastroenterology” in 2002 developed a set of consensus definitions, which has led to the classification of HE into three different types, A-C. (Table 1)[2]. In addition, the clinical presentation of HE was categorised into four main subtypes (Table 2). The heterogeneous nature of the presentation of HE has been the cause of great consternation, and has made the interpretation of comparative studies problematic. The staging of overt HE remains an imprecise art, which is often hampered by its fluctuant course. Thus, more objective methods using electroencephalographic techniques have been developed to assess HE. The effectiveness of using the bispectral index to grade and monitor the course of HE has high discriminative power in patients with both low and high grades of HE, and can be utilised as a simple and objective method of grading HE[7]. It has recently been suggested that we should consider HE as a spectrum of neurocognitive impairment in patients with cirrhosis; the spectrum spanning normal performance to coma[8].

THE AMMONIA HYPOTHESIS

Ammonia was first thought to play a major role in the development of HE when studies by Hahn et al[9], Nencki et al[10] and Nencki et al[11] in the 1890s described the “meat intoxification syndrome”. By diverting blood away from the liver utilising a surgical shunt from the portal vein into the vena cava of dogs, within 6 wk of the portocaval shunt being constructed, it was observed that the dogs developed symptoms such as aggression, irritability, and convulsions, similar to the symptoms exhibited by patients with cirrhosis and overt HE. The portocaval shunt allows blood to bypass the liver, resulting in a lack of urea metabolism, and arterial ammonia levels were found to be increased. When ammonium salts were administered to the dogs, they rapidly fell into a coma and died[9]. Ammonia was later confirmed as the main causative factor of the “meat intoxification syndrome” in portocaval shunted dogs in 1922[12]. The role of ammonia became increasingly recognised as being important when Gabuzda et al[13] and Phillips et al[14] attempted to treat patients with ascites with cation exchange resins that absorbed sodium but released ammonium ions, leading to the adverse effect of significant reversible neurological dysfunction, which was indistinguishable from the syndrome we now know as HE. Blood ammonia concentration was subsequently noted to be elevated in patients with liver disease and hepatic coma[15]; the highest values being found in those patients who were comatose[16].

Subsequently, other investigators have shown that ammonia plays a definitive role in the development of HE. Bessman et al[17] demonstrated a positive arteriovenous difference in ammonia levels in patients with cirrhosis, suggesting an uptake of free ammonia into the brain. More recently, Ehrlich et al[18] demonstrated that by constructing an end-to-side portocaval anastomosis in rats and injecting them with ammonium acetate, the rats demonstrated typical characteristics of HE, such as drowsiness, seizures, and coma in association with elevated blood and brain ammonia concentrations, compared to control rats. Lockwood et al[19] were then able to demonstrate the first evidence linking ammonia to HE in humans, using positron emission tomography (PET). A 13N tracer demonstrated that the rate of uptake of ammonia in the brains of patients was greater in those with HE than without. It was postulated that an increased ammonia uptake in the brain was linked to an increased permeability of the blood-brain barrier to ammonia[20]. In acute liver failure, arterial ammonia concentrations of > 150 mmol/L predict a greater likelihood of dying from brain herniation[21], and intracranial hypertension develops in 55% of cases with an arterial ammonia concentration > 200 mmol/L[1]. In cirrhosis, there is no doubt that blood ammonia concentrations are elevated, but there is conflicting evidence regarding the relationship between ammonia concentration and HE severity. Moreover, it is not unusual in clinical practice to see patients with cirrhosis presenting with symptoms of overt HE who have normal or only mildly elevated arterial ammonia concentrations. Indeed, numerous studies have shown that a single test for blood ammonia concentration is a poor method for assessing HE[22]. Furthermore, Ong et al[23] studied the blood ammonia levels of patients with chronic liver disease and compared these to their mental states. In patients considered not to have any sign of HE, 60% had ammonia levels higher than normal, whereas there was a high proportion of those with grade 3 or 4 HE with normal or only mildly elevated blood ammonia levels. Whilst there is no denying the involvement of ammonia in the pathogenesis of HE, it seems that there might be other factors involved which are as, if not more, important.

THE ASTROCYTE IN HE

Astrocytes are a type of glial cell found within the central nervous system (CNS), which are involved in maintaining cells within the CNS, including providing nutrients for neurones. Astrocytes are particularly vulnerable to the effects of ammonia in the brain. One reason for this is that the enzyme glutamine synthetase is mainly located within astrocytes. Norenberg et al[24] found glutamine synthetase exclusively within astrocytes in rat brains, and none within neurones or other glial cells. It is also important to note that the end-processes of astrocytes surround the capillaries in the CNS. Theoretically, this would ensure that any toxin entering the brain, such as ammonia, is immediately metabolised, protecting other CNS cells from its damaging effects[25]. This theory was tested by Rao et al[26], who investigated the effects of ammonia exposure on purely neuronal cultures and co-cultures of neurons and astrocytes. The cultures containing neurons alone showed significant increases in cell death, apoptotic cells, degeneration of neuronal processes, and free radical levels. However, these changes were not detected in the co-cultures, indicating a protective function of astrocytes.

The blood brain barrier remains anatomically intact in HE[27]; however, PET studies have demonstrated an increased permeability-surface area to ammonia with increasing severity of disease[20].

HE in patients with chronic liver disease is characterised neuropathologically by Alzheimer type Ⅱ astrocytosis. This describes morphological changes to astrocytes, which include a large swollen nucleus, prominent nucleolus, and margination of the chromatin pattern. These neuropathological findings have been replicated in the brains of patients with congenital abnormalities of urea cycle enzymes[28], in experimental animal models[29,30], and astrocyte cultures exposed chronically to ammonia[31]. Therefore, it is likely that ammonia taken up into the brain interacts with astrocytes, eventually leading to these characteristic changes.

In acute liver failure, an increased brain ammonia concentration causes astrocyte swelling and patients develop cytotoxic brain oedema[32]. Kato et al[32] used electron microscopy to study the cells of patients who died of fulminant hepatic failure. They found brain oedema to be present, with pronounced swelling of astrocytes. Glutamine synthetase catalyses the conversion of ammonia and glutamate to glutamine. As a result, hyperammonemia can lead to excessive levels of glutamine within astrocytes, causing the cells to swell, and therefore explaining the oedema and intracranial hypertension seen with fulminant hepatic failure. Willard-Mack et al[33] used rats to investigate whether inducing an acute onset of hyperammonemia caused astrocytes to swell and if inhibiting the action of glutamine synthetase prevented these astrocytic changes. The study found that 8 h after inducing plasma hyperammonemia, changes in astrocyte morphology could be identified. These changes included an increased number of organelles, increased cytoplasmic volume, and an increased nuclear volume. They also found that inhibiting glutamine synthetase attenuated the enlargement of the nuclei and prevented the increase in astrocyte water content seen with hyperammonemia. The results of this study suggest that the production of glutamine by ammonia detoxification, results in water being drawn into astrocytes through osmotic pressure.

There might also be a potential role for vasogenic brain oedema in acute liver failure. This is believed to result from damage to the blood-brain barrier, leading to uncontrolled movement of plasma components and water to extracellular areas of the brain[34]. Consistent with this, animal studies have shown an increased permeability of the blood brain barrier to substances that are normally unable to cross it[35,36]. It has been suggested that perhaps in the early stages of HE, cytotoxic brain oedema predominates, and is enhanced in the later stages by vasogenic brain oedema following damage to the blood brain barrier[37]; however, the ability of mannitol to reduce intracranial hypertension in patients with fulminant hepatic failure indicates that the blood brain barrier remains largely intact[38].

The presence of low-grade astrocyte swelling has been further investigated in human patients with cirrhosis. Córdoba et al[39] used magnetic resonance spectroscopy and the magnetisation transfer ratio (a measure of free water in the brain) to assess cirrhotic patients before and after liver transplantation. The results showed a high level of free water in the brain before liver transplantation, which then reduced after transplantation. This correlated with changes in neuropsychological function, suggesting that brain oedema plays a direct role in the changes observed in HE. A further finding of the study was that brain glutamine levels also correlated with the changes in brain water and neuropsychological function, providing further evidence to the theory that hyperammonemia plays an important role in the pathophysiology of HE. Balata et al[40] showed that inducing hyperammonemia in patients with cirrhosis leads to an increase in brain glutamine, which results in an increase in brain water, and deterioration in neuropsychological function.

Interestingly, brain oedema and the consequent risk of intracranial hypertension are rarely complications of chronic liver failure, and are more often associated with fulminant hepatic failure. One possible suggestion for this is that in chronic liver disease, cells have more time to use compensatory mechanisms to adapt to the osmotic changes taking place[41].

Ammonia is directly toxic to the brain, and in acute liver failure causes disarray of inhibitory and excitatory neurotransmission[42], impairs brain energy metabolism[43-46], alters expression of several genes that code for important proteins involved in brain function[47,48], and impairs autoregulation of cerebral blood flow[49]. In patients with cirrhosis, there appears to be a shift in the balance between inhibitory and excitatory neurotransmission towards a net increase in inhibitory neurotransmission.

THE CHANGING FACE OF HE

Although it is widely accepted that ammonia has a key role to play in the pathophysiology of HE, the clinical picture is not always so straightforward. Frequently, the arterial concentration of ammonia can be elevated in the absence of symptoms of HE, and the correlation between the severity of HE and ammonia concentration in patients with cirrhosis can be poor. The theory that several factors could contribute together to the clinical picture of HE was first suggested by Zieve et al[50] in 1974, who described the possible synergistic effects of several toxins, with ammonia. Since this first suggestion, it has become increasingly apparent that aspects of the inflammatory response (such as elevation of pro-inflammatory cytokines) in response to infection and/or systemic inflammation, and oxidative stress, participate in a synergistic relationship with ammonia in the pathogenesis of HE[51-53].

THE ROLE OF INFECTION AND INFLAMMATION IN HE

Acute liver failure

Studies in patients with acute liver failure have shown a more rapid progression to severe HE in those patients with evidence of a systemic inflammatory response, supporting a link between inflammation and HE[51]. In addition, in patients with acetaminophen-induced acute liver failure, infection and/or the resulting systemic inflammatory response were shown to be important factors contributing to an increase in the severity of HE[52]. Furthermore, in the advanced stages of HE in acute liver failure, the brain produces a number of pro-inflammatory cytokines such as tumor necrosis factor-a (TNF-a), interleukin (IL)-1b and IL-6[54,55]. This relationship is supported by evidence derived from therapeutic interventions, such as moderate hypothermia, that reduce cerebral oedema by reducing cerebral blood flow and inflammatory responses[56,57].

Cirrhosis

In patients with cirrhosis, there is mounting evidence for the role of inflammation in exacerbating the symptoms of HE, thus reinforcing the potential synergistic effects of ammonia and inflammation. Studies have shown this to be the case in patients with minimal HE, and across the whole spectrum of patients with varying degrees of overt HE (Westhaven grades 0-4)[53,58,59]. A recent study confirmed that the presence and severity of minimal HE in cirrhosis is independent of the severity of liver disease and plasma ammonia concentration, but markers of inflammation are significantly higher in those with minimal HE compared to those without[59]. In a further study, significant deterioration of neuropsychological test scores in patients with cirrhosis following induced hyperammonemia during the inflammatory state, but not after its resolution, suggested that inflammation might be important in modulating the cerebral effect of ammonia in liver disease, supporting an inflammatory hypothesis[53].

Synergy with ammonia

As inflammation, infection, and ammonia have been shown to be important in the pathogenesis of HE in cirrhosis, the question has to be raised as to whether infection and inflammation have a synergistic relationship with ammonia[60]. Marini and Broussard used mice with a deficiency in a critical urea cycle enzyme conferring chronic hyperammonemia, to demonstrate an increased sensitivity to inflammation. Furthermore, the hyperammonemic mice developed longer lasting and stronger cognitive defects when exposed to an inflammatory stimulus[61]. In a bile duct ligated (BDL) rat model, Jover et al[62] fed an ammonia-containing diet for 2 wk following ligation and compared animals sacrificed 7 d later to those fed a normal chow diet. Ammonia-fed BDL rats had increased cerebral ammonia and demonstrated the presence of type Ⅱ Alzheimer astrocytosis analogous to patients with cirrhosis presenting with episodic HE. Both BDL groups had evidence of systemic inflammation, but the ammonia-fed BDL rats had increased brain glutamine, decreased brain myoinositol, and a significant increase in brain water compared to BDL controls, alluding to a potential synergistic relationship between ammonia and systemic inflammation. Wright et al[27] went on to explore the hypothesis that the inflammatory response induced by lipopolysaccharide (LPS) exacerbates brain oedema in BDL rats. LPS administration increased brain water in ammonia-fed, BDL, and sham-operated animals significantly, but this was associated with the progression to pre-coma only in the BDL animals. LPS induced cytotoxic brain swelling, but the anatomical integrity of the blood brain barrier was maintained. There was evidence of brain and systemic inflammation in BDL rats, which was significantly increased in LPS-treated animals. Nitrosation of proteins in the frontal cortex of BDL and LPS-treated animals was demonstrated. These data provide further evidence that in a background of cirrhosis and hyperammonemia, superimposed inflammation has an important role in the development of HE.

The ammonia-induced nitrosation of astrocytic proteins shown by Wright et al[27] has also been demonstrated in isolated astrocytes and astroglial tissue in brain sections of portocaval shunted rats[63]. However, ammonia alone cannot be responsible, because protein nitrosation was not demonstrated in ammonia fed sham-operated and ammonia-fed BDL rats in the absence of an inflammatory stimulus. Therefore, both ammonia and an additional inflammatory insult might need to be present for nitrosation of brain proteins to occur in animals with “subliminal” inflammation, such as that which has been observed in the BDL model[27]. This is further supported by recent work that demonstrated the presence of tyrosine nitration in astrocyte cultures in the presence of concentrations of TNF-a typically observed in patients with acute liver failure[64].

Inflammation and the brain

During an episode of infection, cytokines cannot directly cross the blood brain barrier and are unable to have a direct effect. Nevertheless, the peripheral immune system can still signal the brain to elicit a response during infection and inflammation through the expression of pro-inflammatory cytokines such as IL-1b, TNF-a and IL-6, both in the periphery and in the brain. Brain signalling may occur by direct transport of the cytokine across the blood brain barrier via an active transport mechanism, the interaction of the cytokine with circumventricular organs and activation of afferent neurons of the vagus nerve[65]. Endothelial cells, along with the astrocyte, are major constituents of the blood brain barrier. Endothelial cells are activated during infection, resulting in the release of various mediators into the brain. Activated microglial cells and astrocytes have the ability to produce a full repertoire of cytokines in response to inflammation and injury. One such cytokine is IL-1b, which has been shown in vitro to compromise the integrity of the blood brain barrier. This is mediated through the cyclo-oxygenase (COX) pathway within the endothelial cell[66]. In a portocaval shunted rat model that is more akin to a model of minimal HE, Cauli et al[67] demonstrated an improved learning ability following the administration of supra-therapeutic doses of the non-steroidal anti-inflammatory drug (NSAID), ibuprofen. This was accompanied by normalisation of COX and inducible NO activity within the cerebral cortex but interestingly also an increase in TNF-a. It is unclear however, how this NSAID specifically interacts with the glutamate-nitric oxide-cGMP pathway and how COX plays a role in the pathogenesis of minimal HE without identification of the specific COX isoform involved and in the absence of neuroanatomical, proteomic and genomic data. Nevertheless, the therapeutic use of NSAID in HE is not novel. Indomethacin (non-selective COX inhibitor) has been shown in patients with acute liver failure[68], and in a portocaval shunted rat model[69], to improve intracranial hypertension and cerebral oedema. Unfortunately, NSAID use is associated with a number of systemic complications, including cardiovascular/renal compromise and cellular prostaglandin metabolism, which impact greatly on not only astroglial function, but also on the development of organ dysfunction, particularly in the context of patients with longstanding liver disease.

TNF-a is released early during infection and can also influence the permeability of the blood brain barrier[70]. Moreover, an association between circulating TNF-a levels in patients with acute[71] and chronic liver failure[72] and the severity of HE, regardless of aetiology, has been recognised. Endothelial cells have receptors for IL-1b and TNF-a which can transduce signals which ultimately culminate in the intracerebral synthesis of NO and prostanoids[73]. Bémeur et al[74] investigated the effect of IL-1b, TNF-a and interferon-a (IFN-a) gene deletions on the onset of HE. Deletion of the IFN-g gene had no effect on brain water levels or neuropsychiatric status. On the other hand, IL-1b and TNF-a gene deletions significantly delayed the onset of HE and brain oedema.

The relationship between the brain and inflammation is not one way. Molecular and neurophysiological studies during the past decade have suggested that pro-inflammatory responses are controlled by evolutionary neural circuits that operate reflexively[75,76]. The afferent arc of the reflex consists of nerves that sense injury and infection. This activates efferent neural circuits including the cholinergic anti-inflammatory pathway, which modulate immune responses and the progression of inflammatory disease. It might therefore be possible to target neural networks for the treatment of inflammation. This novel and fascinating body of work has recently been reviewed by Tracey[77].

Innate immune dysfunction

Innate immune dysfunction occurs in both acute and chronic liver failure, and up to 50% of admissions to hospital in patients with cirrhosis are likely to be related to the development of infection. In response to infection, the body initiates the innate immune response with phagocytic cells, such as monocytes and neutrophils. This response is particularly relevant to the liver, as the liver is the first organ to encounter bacteria or other toxins absorbed in the gut from the portal vein. Bacterial translocation of organisms from the gut in patients with cirrhosis and portal hypertension results in chronic endotoxemia. This culminates in a local milieu of pro-inflammatory cytokines/chemokines which can upregulate adhesion receptors and activate neutrophils[78]. There is significant literature on the immune response to infection in liver disease, which involves an important role of phagocytes and release of inflammatory cytokines. Patients with cirrhosis are functionally immunosuppressed and have impairment of several host defence mechanisms. The hemodynamic derangement of cirrhosis resembles that produced by endotoxin, and bacteremia can greatly exacerbate this state[79].

Neutrophils are a key component of the innate immune response. Ammonia has been shown to induce neutrophil dysfunction by inducing cell swelling, impaired phagocytosis, and increased oxidative burst in normal neutrophils ex vivo, in ammonia-fed rats and in patients with cirrhosis given an ammonia load[80]. Not only does this make patients potentially vulnerable to developing bacterial and fungal infections, but induces oxidative stress, and may ultimately culminate in a “sepsis-like” immune paralysis[81] and a reduction in monocyte HLA-DR expression[82].

Oxidative Stress

The evidence for the role of oxidative stress in the pathogenesis of HE is incontrovertible. Animal studies have shown significant reductions in the activities of glutathione peroxidase and superoxide dismutase enzymes, both in the liver and brain of rats exposed to ammonium acetate. Superoxide levels, in submitochondrial particles, were found to be elevated in ammonia-exposed rats[83] and lipid peroxidation has been shown to be increased, further demonstrating that hyperammonemia induces oxidative stress[84].

N-methyl D-aspartate (NMDA) receptors play a key role in the production of free radicals and an NMDA antagonist can prevent the calcium-mediated increase in oxidative stress[85]. In vivo excessive ammonia-induced NMDA receptor activation reduces antioxidant enzyme activity and results in increased production of superoxide anions[86]. It is, however, extremely difficult to differentiate whether it is oxidative stress that influences astrocyte swelling or whether astrocyte swelling itself induces oxidative stress through NMDA receptor and calcium-dependent mechanisms[87]. Either way, whether one considers that “the chicken came before the egg or vice versa”, it would imply that the close relationship between astrocyte swelling and oxidative stress leads to an “auto-amplifying signalling loop” which promotes the development of HE[88] (Figure 1).

The production of reactive oxygen species (ROS) can arise in a number of different ways. Aside from ROS arising from neutrophil activation[80] and local and systemic inflammation/infection, ammonia and hypo-osmotic swelling-induced nitric oxide synthesis, the activation of NADPH oxidase[89], and mitochondrial glutamine uptake all generate ROS[90-92]. From these data we can propose a “two-hit hypothesis” in the pathogenesis of HE. Liver dysfunction leads invariably to hyperammonemia, which leads to astrocyte swelling, and in the longer term, structural changes to astrocytes (Alzheimer’s type Ⅱ astrocytosis). After this initial “hit”, a second “hit”, such as an ammonia load following an upper gastrointestinal bleed, systemic inflammation/infection, or the development of hyponatremia in a patient with cirrhosis can drive further astrocyte swelling, oxidative stress, and lead to a rapid deterioration in neuropsychological function (Figure 1 and Table 3).

Uptake of ammonia by astrocytes leads to the production of glutamine through the action of glutamine synthetase. Glutamine exposure in cultured astrocytes increases oxidative stress[91]. Mitochondrial glutamine uptake and subsequent cleavage of glutamine by phosphate-activated glutaminase elevates mitochondrial ammonia, which stimulates ROS production via induction of the mitochondrial permeability transition (MPT)[93]. However, cultured astrocytes exposed to ammonia produce ROS and begin swelling almost immediately, whereas MPT induction and glutamine accumulation occur thereafter.

Although astrocytes are relatively resistant to oxidative and nitrosative stress, neighbouring neurones are vulnerable to free radical attack. This can compromise brain energy metabolism and neurotransmission in patients with HE. Furthermore, ammonia, TNF-a, benzodiazepines, and hyponatremia can all trigger nitric oxide-dependent mobilisation of zinc which can augment GABAergic neurotransmission[94].

The mechanism through which free radical production is increased is currently not fully understood. One suggestion is based on findings that link an increase in calcium release to hyperammonemia. Rose et al[95] exposed cultured mice astrocytes to ammonium chloride. They observed a transient increase in the concentration of calcium ions from intracellular stores. The use of a calcium chelator (BAPTA) prevented the ammonia-induced production of free radicals[25]. Another possibility is that ROS are produced through activation of NMDA receptors[96].

One other area of research interest involves oxidation of RNA. It has been shown that in patients with Alzheimer’s disease, there is significant RNA oxidation, which might result in impairments in protein synthesis and, consequently, cognitive function in patients[97]. Görg et al[98] reported the effects on cultured rat astrocytes and rat brain in vivo of ammonia exposure. Ammonia exposure was associated with a rapid, reversible oxidation of RNA (thought to involve NMDA receptor activation and calcium release). Consistent with this theory is the fact that some substrates required for learning and memory require protein synthesis[96]. Disruption of this protein synthesis via RNA oxidation might therefore interfere with cognitive function.

Therapeutic Strategies in HE

To date, most therapeutic strategies in HE have been focused on lowering arterial concentrations of ammonia and modulating inter-organ ammonia metabolism, but these remain largely ineffective. Treatments based on the hypothesis that the colon is the primary organ responsible for the generation of ammonia have ranged from dietary protein restriction, to the use of non-absorbable disaccharides, non-absorbable antibiotics, and colectomy[99]. However, Córdoba et al[100] showed that diets with normal protein content can be administered safely to patients with cirrhosis with episodic HE and that protein restriction does not have any beneficial effect for cirrhotic patients during an episode of HE and indeed, might even be detrimental in a patient with an underlying catabolic state.

It has been demonstrated that lactulose administered to patients with minimal HE in an unblinded open label study[101] might be of benefit and another open label randomised placebo controlled study in patients with a previous history of overt HE suggested that lactulose might delay the onset of a recurrent episode of HE[102]. However, in a recently published systematic review[103], which had very few high quality studies to base its findings on, lactulose was not found to have any impact on mortality in patients with cirrhosis presenting acutely with overt HE.
The use of non-absorbable antibiotics had been largely abandoned after concern that long-term administration of neomycin might lead to problems with nephrotoxicity and ototoxicity, and with metronidazole might lead to peripheral neuropathy. However, support for this strategy has been recently reinvigorated with the publication of the largest double blind placebo controlled study (n = 299) by Bass et al[104], which compared rifaximin (which has no known long term toxicity) favourably with placebo for the secondary prophylaxis of HE.

Benzodiazepine antagonists such as flumazenil also emerged as a potential therapy for HE patients. An analysis of six randomised controlled trials showed that 27% patients treated with flumazenil showed a clinical improvement, whilst 19% of treated patients showed an electroencephalographic improvement[105].

In the sickest cohorts, direct ammonia removal by hemofiltration in the intensive care unit is effective, but unfortunately by this stage multiorgan dysfunction and bacteremia might have superseded. Likewise, albumin dialysis in patients with acute-on-chronic liver failure improves HE grade[106], but the improvement is independent of changes in ammonia or cytokines[107] and remains controversial[108].

To address the issue of inter-organ ammonia metabolism, recent studies in patients with cirrhosis have shown that other than the gut, kidneys and muscle might be important targets[99]. Volume expansion produces significant increases in renal ammonia excretion resulting in a reduction in plasma ammonia concentration. This was shown to improve mental state, supporting the notion that the kidneys can be manipulated favourably[109]. During the hyperammonemic state, muscle detoxifies ammonia through conversion to glutamine[110,111]. L-ornithine L-aspartate (LOLA), which is a mixture of two amino acids, provides intermediates that increase glutamate availability for synthesis of glutamine and illustrates the concept that muscle can detoxify ammonia. Administration to animals with acute liver failure resulted in reduced brain water[112], but a recent study in patients with acute liver failure did not have any impact on brain dysfunction or survival[113]. When given to patients with cirrhosis and HE, administration of LOLA resulted in an improvement in HE compared with placebo-treated controls[114], although a recent meta-analysis concluded that it had little effect in patients with minimal HE[115]. Jalan et al[116] have hypothesised that this inefficacy might result from an accumulation of glutamine resulting in a rebound rise in circulating ammonia. By utilising a strategy which enables the excretion of glutamine, Davies et al[117] have demonstrated a synergy between L-ornithine and phenylacetate in reducing arterial ammonia in BDL rats.

However, in this review we have already convincingly demonstrated that ammonia, although central in the development of HE, is not solely responsible for its development. Infection/inflammation and oxidative stress are key determinants and indeed act synergistically with ammonia. Although ammonia could potentially be responsible for the development of neutrophil dysfunction, a patient with cirrhosis presents independently as a model of chronic endotoxemia that has direct implications on the innate and adaptive immune systems. We must therefore also look to therapies that directly or indirectly target the proinflammatory milieu.

Potential therapeutic strategies might include NMDA antagonists[84], leukodepletion[118], antagonism of pro-inflammatory cytokines[119], antioxidants [N-acetylcysteine (NAC)[120] and albumin[107,121]], anti-inflammatories (COX inhibitors[67] and minocycline[122,123]), probiotics[124] and hypothermia[125]. Excitement surrounds the prospect of small molecules that modulate toll-like receptor (TLR)-4 signalling, which can potentially down regulate neutrophil activation and other cellular responses. Early data indicate that TLR-4 antagonists can reduce LPS-stimulated cytokine release in healthy volunteers and results from phase 3 clinical trials are awaited. Inhibition of TLR-2, 4, and 9 prevented the increase in neutrophil oxidative burst induced from plasma from patients with alcoholic hepatitis. Furthermore, albumin, an endotoxin scavenger, prevented the deleterious effect of patients’ plasma on neutrophil phagocytosis, spontaneous oxidative burst, and TLR expression[121]. This might also explain the beneficial role of albumin dialysis on HE[107,126].

When administered early after an overdose of acetaminophen, intravenous NAC prevents hepatic necrosis by replenishing stores of glutathione[127]. In patients with acute liver failure secondary to an overdose of acetaminophen, and in patients with acute liver failure secondary to other causes, NAC has been shown to increase oxygen delivery and consumption associated with increases in mean arterial pressure, cardiac index[128], and cerebral perfusion pressure[120]. These beneficial hemodynamic effects have been shown to be mediated by enhanced activity of the nitric oxide/soluble cGMP system[129] and suggest that NAC could have a beneficial role in the treatment of patients with cirrhosis who have developed overt HE.

As the role of central pro-inflammatory mechanisms are believed to be important in the pathogenesis of HE, then another novel therapeutic candidate drug to be considered is minocycline, which has been shown in two very recent studies by Jiang et al[122,123] to have anti-inflammatory effects in rats with acute liver failure. Minocyline treatment prevented both microglial activation [CD11b/c (OX-42) expression on immunohistochemistry] as well as the upregulation of IL-1b, IL-6, TNF-a, heme-oxygenase-1, eNOS, iNOS mRNA and protein expression with a concomitant attenuation of the progression of HE and brain edema, and at least in part, by reduction of oxidative/nitrosative stress. Thus, minocycline might also have promise in patients with acute and chronic liver failure cirrhosis and HE, and could be taken forward into randomised placebo controlled trials.

Modulation of intestinal microbiota is an emerging strategy to reduce the bacterial translocation of LPS and other bacterial activators of TLRs. Probiotics have been shown to reduce bacterial translocation and were shown to improve liver function and prevent the development of infection and HE in patients with cirrhosis[124]. Furthermore, probiotics have been shown to restore neutrophil phagocytic capacity in patients with alcoholic cirrhosis, possibly by reducing endogenous levels of IL-10 and TLR-4 expression[130].

Recent studies show that hypothermia is efficacious in patients with uncontrolled intracranial hypertension that are undergoing liver transplantation[56,125]. Hypothermia displays many beneficial effects on brain water and intracranial hypertension relating to decreased brain ammonia, cerebral blood flow, mediators of inflammation, and oxidative stress[131]. The sites of action of potential therapies for HE is shown in Figure 1.

CONCLUSION

HE is a dynamic neuropsychological spectral disorder that develops after liver injury. The pathophysiological mechanisms behind the development of HE are still not fully understood, but ammonia and the downstream consequences of ammonia uptake by astrocytes remain fundamental to the process. Ammonia not only leads to astrocyte swelling, but also alters neurotransmission, mitochondrial function, and induces oxidative stress. Astrocyte swelling and oxidative stress are closely related and result in “an auto-amplifying” loop. The presence of local and systemic inflammation and the release of ROS further exacerbate the cerebral effects of ammonia. Anti-inflammatory and anti-oxidative strategies may abrogate these effects and offer real treatment options to patients with HE in the future.

REFERENCES

1 Bernal W, Hall C, Karvellas CJ, Auzinger G, Sizer E, Wendon J. Arterial ammonia and clinical risk factors for encephalopathy and intracranial hypertension in acute liver failure. Hepatology 2007; 46: 1844-1852

2 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

3 Schomerus H, Hamster W. Quality of life in cirrhotics with minimal hepatic encephalopathy. Metab Brain Dis 2001; 16: 37-41

4 Schomerus H, Hamster W, Blunck H, Reinhard U, Mayer K, Dölle W. Latent portasystemic encephalopathy. I. Nature of cerebral functional defects and their effect on fitness to drive. Dig Dis Sci 1981; 26: 622-630

5 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

6 Mechtcheriakov S, Graziadei IW, Mattedi M, Bodner T, Kugener A, Hinterhuber HH, Marksteiner J, Vogel W. Incomplete improvement of visuo-motor deficits in patients with minimal hepatic encephalopathy after liver transplantation. Liver Transpl 2004; 10: 77-83

7 Dahaba AA, Worm HC, Zhu SM, Bao FP, Salah A, Zakaria S, Bornemann H, Stadlbauer V, Rehak PH, Metzler H, Stauber RE. Sensitivity and specificity of bispectral index for classification of overt hepatic encephalopathy: a multicentre, observer blinded, validation study. Gut 2008; 57: 77-83

8 Bajaj JS, Wade JB, Sanyal AJ. Spectrum of neurocognitive impairment in cirrhosis: Implications for the assessment of hepatic encephalopathy. Hepatology 2009; 50: 2014-2021

9 Hahn M, Massen O, Nencki M, Pawlow I. Eck's fistula between the inferior vena cava and the portal vein: its consequences for the organism. Naunyn Schmiedebergs Arch Pharmacol 1893; 32: 161-210

10 Nencki M, Zaleski J. On the quantitation of ammonia in animal fluids and tissues. Naunyn Schmiedebergs Arch Exp Pathol Pharmakol 1895; 36: 385-396

11 Nencki M, Pawlow J, Zaleski J. On the content of ammonia in blood within organs and on formation of urea in mammals. Naunyn Schmiedebergs Arch Exp Pathol Pharmakol 1896; 37: 26-51

12 Matthews SA. Ammonia, a causative factor in meat poison- ing in Eck fistula dogs. Am J Physiol 1922; 59: 459-460

13 Gabuzda GJ Jr, Phillips GB, Davidson CS. Reversible toxic manifestations in patients with cirrhosis of the liver given cation-exchange resins. N Engl J Med 1952; 246: 124-130

14 Phillips GB, Schwartz R, Gabuzda GJ Jr, Davidson CS. The syndrome of impending hepatic coma in patients with cirrhosis of the liver given certain nitrogenous substances. N Engl J Med 1952; 247: 239-246

15 Traeger HS, Gabuzda GJ Jr, Ballou AN, Davidson CS. Blood ammonia concentration in liver disease, and liver coma. Metabolism 1954; 3: 99-109

16 Phear EA, Sherlock S, Summerskill WH. Blood-ammonium levels in liver disease and hepatic coma. Lancet 1955; 268: 836-840

17 Bessman SP, Bessman AN. The cerebral and peripheral uptake of ammonia in liver disease with an hypothesis for the mechanism of hepatic coma. J Clin Invest 1955; 34: 622-628

18 Ehrlich M, Plum F, Duffy TE. Blood and brain ammonia concentrations after portacaval anastomosis. Effects of acute ammonia loading. J Neurochem 1980; 34: 1538-1542

19 Lockwood AH, McDonald JM, Reiman RE, Gelbard AS, Laughlin JS, Duffy TE, Plum F. The dynamics of ammonia metabolism in man. Effects of liver disease and hyperammonemia. J Clin Invest 1979; 63: 449-460

20 Lockwood AH, Yap EW, Wong WH. Cerebral ammonia metabolism in patients with severe liver disease and minimal hepatic encephalopathy. J Cereb Blood Flow Metab 1991; 11: 337-341

21 Clemmesen JO, Larsen FS, Kondrup J, Hansen BA, Ott P. Cerebral herniation in patients with acute liver failure is correlated with arterial ammonia concentration. Hepatology 1999; 29: 648-653

22 Arora S, Martin CL, Herbert M. Myth: interpretation of a single ammonia level in patients with chronic liver disease can confirm or rule out hepatic encephalopathy. CJEM 2006; 8: 433-435

23 Ong JP, Aggarwal A, Krieger D, Easley KA, Karafa MT, Van Lente F, Arroliga AC, Mullen KD. Correlation between ammonia levels and the severity of hepatic encephalopathy. Am J Med 2003; 114: 188-193

24 Norenberg MD, Martinez-Hernandez A. Fine structural localization of glutamine synthetase in astrocytes of rat brain. Brain Res 1979; 161: 303-310

25 Norenberg MD, Rama Rao KV, Jayakumar AR. Signaling factors in the mechanism of ammonia neurotoxicity. Metab Brain Dis 2009; 24: 103-117

26 Rao KV, Panickar KS, Jayakumar AR, Norenberg MD. Astrocytes protect neurons from ammonia toxicity. Neurochem Res 2005; 30: 1311-1318

27 Wright G, Davies NA, Shawcross DL, Hodges SJ, Zwingmann C, Brooks HF, Mani AR, Harry D, Stadlbauer V, Zou Z, Williams R, Davies C, Moore KP, Jalan R. Endotoxemia produces coma and brain swelling in bile duct ligated rats. Hepatology 2007; 45: 1517-1526

28 Plum F, Hindfelt B. The neurological complications of liver disease. In: Vinken P, Bruyn G, editors. Handbook of clinical neurology. Volume 27. New York: American Elsevier, 1976: 349-377

29 Gibson GE, Zimber A, Krook L, Richardson EP, Visek WJ. Brain histology and behavior of mice injected with urease. J Neuropathol Exp Neurol 1974; 33: 201-211

30 Norenberg MD. A light and electron microscopic study of experimental portal-systemic (ammonia) encephalopathy. Progression and reversal of the disorder. Lab Invest 1977; 36: 618-627

31 Norenberg M. Hepatic encephalopathy: studies with astrocyte cultures. In: Norenberg M, Hertz L, Schousboe A, editors. The biochemical pathology of astrocytes. New York: Alan R. Liss, 1988: 451-464

32 Kato M, Hughes RD, Keays RT, Williams R. Electron microscopic study of brain capillaries in cerebral edema from fulminant hepatic failure. Hepatology 1992; 15: 1060-1066

33 Willard-Mack CL, Koehler RC, Hirata T, Cork LC, Takahashi H, Traystman RJ, Brusilow SW. Inhibition of glutamine synthetase reduces ammonia-induced astrocyte swelling in rat. Neuroscience 1996; 71: 589-599

34 Larsen FS. Cerebral circulation in liver failure: Ohm's law in force. Semin Liver Dis 1996; 16: 281-292

35 Dixit V, Chang TM. Brain edema and the blood brain barrier in galactosamine-induced fulminant hepatic failure rats. An animal model for evaluation of liver support systems. ASAIO Trans 1990; 36: 21-27

36 Horowitz ME, Schafer DF, Molnar P, Jones EA, Blasberg RG, Patlak CS, Waggoner J, Fenstermacher JD. Increased blood-brain transfer in a rabbit model of acute liver failure. Gastroenterology 1983; 84: 1003-1011

37 Detry O, De Roover A, Honore P, Meurisse M. Brain edema and intracranial hypertension in fulminant hepatic failure: pathophysiology and management. World J Gastroenterol 2006; 12: 7405-7412

38 Canalese J, Gimson AE, Davis C, Mellon PJ, Davis M, Williams R. Controlled trial of dexamethasone and mannitol for the cerebral oedema of fulminant hepatic failure. Gut 1982; 23: 625-629

39 Córdoba J, Alonso J, Rovira A, Jacas C, Sanpedro F, Castells L, Vargas V, Margarit C, Kulisewsky J, Esteban R, Guardia J. The development of low-grade cerebral edema in cirrhosis is supported by the evolution of (1)H-magnetic resonance abnormalities after liver transplantation. J Hepatol 2001; 35: 598-604

40 Balata S, Olde Damink SW, Ferguson K, Marshall I, Hayes PC, Deutz NE, Williams R, Wardlaw J, Jalan R. Induced hyperammonemia alters neuropsychology, brain MR spectroscopy and magnetization transfer in cirrhosis. Hepatology 2003; 37: 931-939

41 Shawcross DL, Balata S, Olde Damink SW, Hayes PC, Wardlaw J, Marshall I, Deutz NE, Williams R, Jalan R. Low myo-inositol and high glutamine levels in brain are associated with neuropsychological deterioration after induced hyperammonemia. Am J Physiol Gastrointest Liver Physiol 2004; 287: G503-G509

42 Szerb JC, Butterworth RF. Effect of ammonium ions on synaptic transmission in the mammalian central nervous system. Prog Neurobiol 1992; 39: 135-153

43 Lai JC, Cooper AJ. Brain alpha-ketoglutarate dehydrogenase complex: kinetic properties, regional distribution, and effects of inhibitors. J Neurochem 1986; 47: 1376-1386

44 Hindfelt B, Plum F, Duffy TE. Effect of acute ammonia intoxication on cerebral metabolism in rats with portacaval shunts. J Clin Invest 1977; 59: 386-396

45 Yao H, Sadoshima S, Fujii K, Kusuda K, Ishitsuka T, Tamaki K, Fujishima M. Cerebrospinal fluid lactate in patients with hepatic encephalopathy. Eur Neurol 1987; 27: 182-187

46 Zwingmann C, Chatauret N, Leibfritz D, Butterworth RF. Selective increase of brain lactate synthesis in experimental acute liver failure: results of a [H-C] nuclear magnetic resonance study. Hepatology 2003; 37: 420-428

47 Desjardins P, Bélanger M, Butterworth RF. Alterations in expression of genes coding for key astrocytic proteins in acute liver failure. J Neurosci Res 2001; 66: 967-971

48 Rama Rao KV, Chen M, Simard JM, Norenberg MD. Increased aquaporin-4 expression in ammonia-treated cultured astrocytes. Neuroreport 2003; 14: 2379-2382

49 Larsen FS, Strauss G, Møller K, Hansen BA. Regional cerebral blood flow autoregulation in patients with fulminant hepatic failure. Liver Transpl 2000; 6: 795-800

50 Zieve L, Doizaki WM, Zieve J. Synergism between mercaptans and ammonia or fatty acids in the production of coma: a possible role for mercaptans in the pathogenesis of hepatic coma. J Lab Clin Med 1974; 83: 16-28
51 Rolando N, Wade J, Davalos M, Wendon J, Philpott-Howard J, Williams R. The systemic inflammatory response syndrome in acute liver failure. Hepatology 2000; 32: 734-739

52 Vaquero J, Polson J, Chung C, Helenowski I, Schiodt FV, Reisch J, Lee WM, Blei AT. Infection and the progression of hepatic encephalopathy in acute liver failure. Gastroenterology 2003; 125: 755-764

53 Shawcross DL, Davies NA, Williams R, Jalan R. Systemic inflammatory response exacerbates the neuropsychological effects of induced hyperammonemia in cirrhosis. J Hepatol 2004; 40: 247-254

54 Jalan R, Olde Damink SWM, Lee A, Hayes PC, Williams R. Brain production of pro-inflammatory cytokines in patients with acute liver failure and uncontrolled intracranial hypertension: evidence of disrupted blood brain barrier [Abstract]? Hepatology 2003; 38: 548

55 Wright G, Shawcross D, Olde Damink SW, Jalan R. Brain cytokine flux in acute liver failure and its relationship with intracranial hypertension. Metab Brain Dis 2007; 22: 375-388

56 Jalan R, Olde Damink SW, Deutz NE, Davies NA, Garden OJ, Madhavan KK, Hayes PC, Lee A. Moderate hypothermia prevents cerebral hyperemia and increase in intracranial pressure in patients undergoing liver transplantation for acute liver failure. Transplantation 2003; 75: 2034-2039

57 Jalan R, Pollok A, Shah SH, Madhavan K, Simpson KJ. Liver derived pro-inflammatory cytokines may be important in producing intracranial hypertension in acute liver failure. J Hepatol 2002; 37: 536-538

58 Sharifi Y, Yeoman A, Austin M, Abeles R, Portal A, Sizer E, Auzinger G, Bernal W, Wendon J, Shawcross D. Defining the outcomes of severe hepatic encephalopathy in cirrhosis: inflammation is the key. Hepatology 2008; 48: 1061A

59 Shawcross DL, Wright G, Olde Damink SW, Jalan R. Role of ammonia and inflammation in minimal hepatic encephalopathy. Metab Brain Dis 2007; 22: 125-138

60 Blei AT. Infection, inflammation and hepatic encephalopathy, synergism redefined. J Hepatol 2004; 40: 327-330

61 Marini JC, Broussard SR. Hyperammonemia increases sensitivity to LPS. Mol Genet Metab 2006; 88: 131-137

62 Jover R, Rodrigo R, Felipo V, Insausti R, Sáez-Valero J, García-Ayllón MS, Suárez I, Candela A, Compañ A, Esteban A, Cauli O, Ausó E, Rodríguez E, Gutiérrez A, Girona E, Erceg S, Berbel P, Pérez-Mateo M. Brain edema and inflammatory activation in bile duct ligated rats with diet-induced hyperammonemia: A model of hepatic encephalopathy in cirrhosis. Hepatology 2006; 43: 1257-1266

63 Schliess F, Görg B, Fischer R, Desjardins P, Bidmon HJ, Herrmann A, Butterworth RF, Zilles K, Häussinger D. Ammonia induces MK-801-sensitive nitration and phosphorylation of protein tyrosine residues in rat astrocytes. FASEB J 2002; 16: 739-741

64 Görg B, Bidmon HJ, Keitel V, Foster N, Goerlich R, Schliess F, Häussinger D. Inflammatory cytokines induce protein tyrosine nitration in rat astrocytes. Arch Biochem Biophys 2006; 449: 104-114

65 Licinio J, Wong ML. Pathways and mechanisms for cytokine signaling of the central nervous system. J Clin Invest 1997; 100: 2941-2947

66 de Vries HE, Blom-Roosemalen MC, van Oosten M, de Boer AG, van Berkel TJ, Breimer DD, Kuiper J. The influence of cytokines on the integrity of the blood-brain barrier in vitro. J Neuroimmunol 1996; 64: 37-43

67 Cauli O, Rodrigo R, Piedrafita B, Boix J, Felipo V. Inflammation and hepatic encephalopathy: ibuprofen restores learning ability in rats with portacaval shunts. Hepatology 2007; 46: 514-519

68 Tofteng F, Larsen FS. The effect of indomethacin on intracranial pressure, cerebral perfusion and extracellular lactate and glutamate concentrations in patients with fulminant hepatic failure. J Cereb Blood Flow Metab 2004; 24: 798-804

69 Chung C, Gottstein J, Blei AT. Indomethacin prevents the development of experimental ammonia-induced brain edema in rats after portacaval anastomosis. Hepatology 2001; 34: 249-254

70 Didier N, Romero IA, Créminon C, Wijkhuisen A, Grassi J, Mabondzo A. Secretion of interleukin-1beta by astrocytes mediates endothelin-1 and tumour necrosis factor-alpha effects on human brain microvascular endothelial cell permeability. J Neurochem 2003; 86: 246-254

71 Streetz K, Leifeld L, Grundmann D, Ramakers J, Eckert K, Spengler U, Brenner D, Manns M, Trautwein C. Tumor necrosis factor alpha in the pathogenesis of human and murine fulminant hepatic failure. Gastroenterology 2000; 119: 446-460

72 Odeh M, Sabo E, Srugo I, Oliven A. Serum levels of tumor necrosis factor-alpha correlate with severity of hepatic encephalopathy due to chronic liver failure. Liver Int 2004; 24: 110-116

73 Schiltz JC, Sawchenko PE. Signaling the brain in systemic inflammation: the role of perivascular cells. Front Biosci 2003; 8: s1321-s1329

74 Bémeur C, Qu H, Desjardins P, Butterworth RF. IL-1 or TNF receptor gene deletion delays onset of encephalopathy and attenuates brain edema in experimental acute liver failure. Neurochem Int 2010; 56: 213-215

75 Tracey KJ. The inflammatory reflex. Nature 2002; 420: 853-859

76 Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, Wang H, Abumrad N, Eaton JW, Tracey KJ. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 2000; 405: 458-462

77 Tracey KJ. Reflex control of immunity. Nat Rev Immunol 2009; 9: 418-428

78 Bode C, Kugler V, Bode JC. Endotoxemia in patients with alcoholic and non-alcoholic cirrhosis and in subjects with no evidence of chronic liver disease following acute alcohol excess. J Hepatol 1987; 4: 8-14

79 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

80 Shawcross DL, Wright GA, Stadlbauer V, Hodges SJ, Davies NA, Wheeler-Jones C, Pitsillides AA, Jalan R. Ammonia impairs neutrophil phagocytic function in liver disease. Hepatology 2008; 48: 1202-1212

81 Wasmuth HE, Kunz D, Yagmur E, Timmer-Stranghöner A, Vidacek D, Siewert E, Bach J, Geier A, Purucker EA, Gressner AM, Matern S, Lammert F. Patients with acute on chronic liver failure display "sepsis-like" immune paralysis. J Hepatol 2005; 42: 195-201

82 Antoniades CG, Wendon J, Vergani D. Paralysed monocytes in acute on chronic liver disease. J Hepatol 2005; 42: 163-165

83 Kosenko E, Kaminsky Y, Kaminsky A, Valencia M, Lee L, Hermenegildo C, Felipo V. Superoxide production and antioxidant enzymes in ammonia intoxication in rats. Free Radic Res 1997; 27: 637-644

84 Kosenko E, Kaminski Y, Lopata O, Muravyov N, Felipo V. Blocking NMDA receptors prevents the oxidative stress induced by acute ammonia intoxication. Free Radic Biol Med 1999; 26: 1369-1374

85 Mattson MP, Lovell MA, Furukawa K, Markesbery WR. Neurotrophic factors attenuate glutamate-induced accumulation of peroxides, elevation of intracellular Ca2+ concentration, and neurotoxicity and increase antioxidant enzyme activities in hippocampal neurons. J Neurochem 1995; 65: 1740-1751

86 Kosenko E, Venediktova N, Kaminsky Y, Montoliu C, Felipo V. Sources of oxygen radicals in brain in acute ammonia intoxication in vivo. Brain Res 2003; 981: 193-200

87 Schliess F, Görg B, Häussinger D. Pathogenetic interplay between osmotic and oxidative stress: the hepatic encephalopathy paradigm. Biol Chem 2006; 387: 1363-1370

88 Häussinger D. Low grade cerebral edema and the pathogenesis of hepatic encephalopathy in cirrhosis. Hepatology 2006; 43: 1187-1190
89 Reinehr R, Görg B, Becker S, Qvartskhava N, Bidmon HJ, Selbach O, Haas HL, Schliess F, Häussinger D. Hypoosmotic swelling and ammonia increase oxidative stress by NADPH oxidase in cultured astrocytes and vital brain slices. Glia 2007; 55: 758-771

90 Jayakumar AR, Rao KV, Murthy ChR, Norenberg MD. Glutamine in the mechanism of ammonia-induced astrocyte swelling. Neurochem Int 2006; 48: 623-628

91 Jayakumar AR, Rama Rao KV, Schousboe A, Norenberg MD. Glutamine-induced free radical production in cultured astrocytes. Glia 2004; 46: 296-301

92 Jayakumar AR, Panickar KS, Murthy ChR, Norenberg MD. Oxidative stress and mitogen-activated protein kinase phosphorylation mediate ammonia-induced cell swelling and glutamate uptake inhibition in cultured astrocytes. J Neurosci 2006; 26: 4774-4784

93 Bai G, Rama Rao KV, Murthy CR, Panickar KS, Jayakumar AR, Norenberg MD. Ammonia induces the mitochondrial permeability transition in primary cultures of rat astrocytes. J Neurosci Res 2001; 66: 981-991

94 Celentano JJ, Gyenes M, Gibbs TT, Farb DH. Negative modulation of the gamma-aminobutyric acid response by extracellular zinc. Mol Pharmacol 1991; 40: 766-773

95 Rose C, Kresse W, Kettenmann H. Acute insult of ammonia leads to calcium-dependent glutamate release from cultured astrocytes, an effect of pH. J Biol Chem 2005; 280: 20937-20944

96 Häussinger D, Görg B. Interaction of oxidative stress, astrocyte swelling and cerebral ammonia toxicity. Curr Opin Clin Nutr Metab Care 2010; 13: 87-92

97 Ding Q, Markesbery WR, Chen Q, Li F, Keller JN. Ribosome dysfunction is an early event in Alzheimer's disease. J Neurosci 2005; 25: 9171-9175

98 Görg B, Qvartskhava N, Keitel V, Bidmon HJ, Selbach O, Schliess F, Häussinger D. Ammonia induces RNA oxidation in cultured astrocytes and brain in vivo. Hepatology 2008; 48: 567-579

99 Shawcross D, Jalan R. Dispelling myths in the treatment of hepatic encephalopathy. Lancet 2005; 365: 431-433

100 Córdoba J, López-Hellín J, Planas M, Sabín P, Sanpedro F, Castro F, Esteban R, Guardia J. Normal protein diet for episodic hepatic encephalopathy: results of a randomized study. J Hepatol 2004; 41: 38-43

101 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

102 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

103 Als-Nielsen B, Gluud LL, Gluud C. Non-absorbable disaccharides for hepatic encephalopathy: systematic review of randomised trials. BMJ 2004; 328: 1046

104 Bass NM, Mullen KD, Sanyal A, Poordad F, Neff G, Leevy CB, Sigal S, Sheikh MY, Beavers K, Frederick T, Teperman L, Hillebrand D, Huang S, Merchant K, Shaw A, Bortey E, Forbes WP. Rifaximin treatment in hepatic encephalopathy. N Engl J Med 2010; 362: 1071-1081

105 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

106 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

107 Sen S, Williams R, Jalan R. The pathophysiological basis of acute-on-chronic liver failure. Liver 2002; 22 Suppl 2: 5-13

108 Ferenci P, Kramer L. MARS and the failing liver-Any help from the outer space? Hepatology 2007; 46: 1682-1684

109 Jalan R, Kapoor D. Reversal of diuretic-induced hepatic encephalopathy with infusion of albumin but not colloid. Clin Sci (Lond) 2004; 106: 467-474

110 Olde Damink SW, Jalan R, Redhead DN, Hayes PC, Deutz NE, Soeters PB. Interorgan ammonia and amino acid metabolism in metabolically stable patients with cirrhosis and a TIPSS. Hepatology 2002; 36: 1163-1171

111 Olde Damink SW, Jalan R, Deutz NE, Redhead DN, Dejong CH, Hynd P, Jalan RA, Hayes PC, Soeters PB. The kidney plays a major role in the hyperammonemia seen after simulated or actual GI bleeding in patients with cirrhosis. Hepatology 2003; 37: 1277-1285

112 Rose C, Michalak A, Rao KV, Quack G, Kircheis G, Butterworth RF. L-ornithine-L-aspartate lowers plasma and cerebrospinal fluid ammonia and prevents brain edema in rats with acute liver failure. Hepatology 1999; 30: 636-640

113 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

114 Kircheis G, Nilius R, Held C, Berndt H, Buchner M, Görtelmeyer R, Hendricks R, Krüger B, Kuklinski B, Meister H, Otto HJ, Rink C, Rösch W, Stauch S. 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

115 Jiang Q, Jiang XH, Zheng MH, Chen YP. L-Ornithine-l-aspartate in the management of hepatic encephalopathy: a meta-analysis. J Gastroenterol Hepatol 2009; 24: 9-14

116 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

117 Davies NA, Wright G, Ytrebø LM, Stadlbauer V, Fuskevåg OM, Zwingmann C, Davies DC, Habtesion A, Hodges SJ, Jalan R. L-ornithine and phenylacetate synergistically produce sustained reduction in ammonia and brain water in cirrhotic rats. Hepatology 2009; 50: 155-164

118 Treacher DF, Sabbato M, Brown KA, Gant V. The effects of leucodepletion in patients who develop the systemic inflammatory response syndrome following cardiopulmonary bypass. Perfusion 2001; 16 Suppl: 67-73

119 Netea MG, van der Meer JW, van Deuren M, Kullberg BJ. Proinflammatory cytokines and sepsis syndrome: not enough, or too much of a good thing? Trends Immunol 2003; 24: 254-258

120 Ytrebø LM, Korvald C, Nedredal GI, Elvenes OP, Nielsen Grymyr OJ, Revhaug A. N-acetylcysteine increases cerebral perfusion pressure in pigs with fulminant hepatic failure. Crit Care Med 2001; 29: 1989-1995

121 Stadlbauer V, Mookerjee RP, Wright GA, Davies NA, Jürgens G, Hallström S, Jalan R. Role of Toll-like receptors 2, 4, and 9 in mediating neutrophil dysfunction in alcoholic hepatitis. Am J Physiol Gastrointest Liver Physiol 2009; 296: G15-G22

122 Jiang W, Desjardins P, Butterworth RF. Cerebral inflammation contributes to encephalopathy and brain edema in acute liver failure: protective effect of minocycline. J Neurochem 2009; 109: 485-493

123 Jiang W, Desjardins P, Butterworth RF. Minocycline attenuates oxidative/nitrosative stress and cerebral complications of acute liver failure in rats. Neurochem Int 2009; 55: 601-605

124 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

125 Jalan R, O Damink SW, Deutz NE, Lee A, Hayes PC. Moderate hypothermia for uncontrolled intracranial hypertension in acute liver failure. Lancet 1999; 354: 1164-1168

126 Stadlbauer V, Krisper P, Aigner R, Haditsch B, Jung A, Lackner C, Stauber RE. Effect of extracorporeal liver support by MARS and Prometheus on serum cytokines in acute-on-chronic liver failure. Crit Care 2006; 10: R169

127 Harrison PM, Keays R, Bray GP, Alexander GJ, Williams R. Improved outcome of paracetamol-induced fulminant hepatic failure by late administration of acetylcysteine. Lancet 1990; 335: 1572-1573

128 Harrison PM, Wendon JA, Gimson AE, Alexander GJ, Williams R. Improvement by acetylcysteine of hemodynamics and oxygen transport in fulminant hepatic failure. N Engl J Med 1991; 324: 1852-1857

129 Harrison P, Wendon J, Williams R. Evidence of increased guanylate cyclase activation by acetylcysteine in fulminant hepatic failure. Hepatology 1996; 23: 1067-1072

130 Stadlbauer V, Mookerjee RP, Hodges S, Wright GA, Davies NA, Jalan R. Effect of probiotic treatment on deranged neutrophil function and cytokine responses in patients with compensated alcoholic cirrhosis. J Hepatol 2008; 48: 945-951

131 Jalan R, Olde Damink SW, Deutz NE, Hayes PC, Lee A. Moderate hypothermia in patients with acute liver failure and uncontrolled intracranial hypertension. Gastroenterology 2004; 127: 1338-1346

Source

Treatment of hepatitis C in HCV mono-infected and in HIV-HCV co-infected patients: an open-labelled comparison study.

Swiss Med Wkly. 2010 Jul 19;140:w13055. doi: 10.4414/smw.2010.13055.

Gonvers JJ, Heim MH, Cavassini M, Müllhaupt B, Genné D, Bernasconi E, Borovicka J, Cerny A, Chave JP, Chuard C, Dufour F, Dutoit V, Malinverni R, Monnat M, Negro F, Troilliet N, Oneta C.

Department of Gastroenterology & Hepatology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland. Jean-Jacques.Gonvers@chuv.ch

Abstract

BACKGROUND/AIMS: Treatment of chronic HCV infection has become a priority in HIV+ patients, given the faster progression to end-stage liver disease. The primary endpoint of this study was to evaluate and compare antiviral efficacy of Peginterferon alpha 2a plus ribavirin in HIV-HCV co-infected and HCV mono-infected patients, and to examine whether 6 months of therapy would have the same efficacy in HIV patients with favourable genotypes 2 and 3 as in mono-infected patients, to minimise HCV-therapy-related toxicities. Secondary endpoints were to evaluate predictors of sustained virological response (SVR) and frequency of side-effects. METHODS: Patients with genotypes 1 and 4 were treated for 48 weeks with Pegasys 180 microg/week plus Copegus 1000-1200 mg/day according to body weight; patients with genotypes 2 and 3 for 24 weeks with Pegasys 180 microg/week plus Copegus 800 mg/day. RESULTS: 132 patients were enrolled in the study: 85 HCV mono-infected (38: genotypes 1 and 4; 47: genotypes 2 and 3), 47 HIV-HCV co-infected patients (23: genotypes 1 and 4; 24: genotypes 2 and 3). In an intention-to-treat analysis, SVR for genotypes 1 and 4 was observed in 58% of HCV mono-infected and in 13% of HIV-HCV co-infected patients (P = 0.001). For genotypes 2 and 3, SVR was observed in 70% of HCV mono-infected and in 67% of HIV-HCV co-infected patients (P = 0.973). Undetectable HCV-RNA at week 4 had a positive predictive value for SVR for mono-infected patients with genotypes 1 and 4 of 0.78 (95% CI: 0.54-0.93) and of 0.81 (95% CI: 0.64-0.92) for genotypes 2 and 3. For co-infected patients with genotypes 2 and 3, the positive predictive value of SVR of undetectable HCV-RNA at week 4 was 0.76 (95%CI, 0.50-0.93). Study not completed by 22 patients (36%): genotypes 1 and 4 and by 12 patients (17%): genotypes 2 and 3. CONCLUSION: Genotypes 2 or 3 predict the likelihood of SVR in HCV mono-infected and in HIV-HCV co-infected patients. A 6-month treatment with Peginterferon alpha 2a plus ribavirin has the same efficacy in HIV-HCV co-infected patients with genotypes 2 and 3 as in mono-infected patients. HCV-RNA negativity at 4 weeks has a positive predictive value for SVR. Aggressive treatment of adverse effects to avoid dose reduction, consent withdrawal or drop-out is crucial to increase the rate of SVR, especially when duration of treatment is 48 weeks. Sixty-one percent of HIV-HCV co-infected patients with genotypes 1 and 4 did not complete the study against 4% with genotypes 2 and 3.

PMID: 20648398 [PubMed - in process]Free Article

Source

Concomitant nevirapine therapy is associated with higher efficacy of pegylated interferon plus ribavirin among HIV/hepatitis C virus-coinfected patients

Reported by Jules Levin

JA Mira, LF Lopez-Cortes, E Vispo, C Tural, P Mallolas, E Ferrer, I de los Santos-Gil, P Domingo, H Knobel, F Tellez, M Crespo, A Rivero, E Ortega, JA Pineda on behalf of the VIRA-C Study Group




























Source

HCV SVR Rate Higher With Nevirapine Than Lopinavir in Multicenter Study

XVIII International AIDS Conference, July 18-23, 2010, Vienna
Mark Mascolini

Compared with people taking a lopinavir (LPV)/ritonavir-based antiretroviral regimen, those taking a nevirapine (NVP)-containing combination were more likely to attain a sustained virologic response (SVR) when treated for hepatitis C virus (HCV) infection with pegylated interferon and ribavirin (PEG-IFN/RBV) [1]. Results must be interpreted cautiously, however, because this is a retrospective cohort study, not a randomized trial, and there were important differences between the lopinavir and nevirapine groups.

Earlier research found that HIV/HCV-coinfected people taking nevirapine had lower HCV loads than people taking an antiretroviral regimen including a protease inhibitor (PI). To see whether nevirapine is also associated with higher SVR rates, Spanish investigators planned this 20-center retrospective study of 71 coinfected adults (43%) taking a nevirapine regimen and 94 (57%) taking lopinavir/ritonavir when they started PEG-IFN/RBV for the first time. Everyone took nevirapine or lopinavir with tenofovir plus either lamivudine or emtricitabine. People took PEG-IFN/RBV for 48 weeks if they had HCV genotype 1 or 4, and for 24 or 48 weeks if they had genotype 2 or 3.

The antiretroviral groups were similar in age (median 42 with nevirapine and 41 with lopinavir), percentage of former injection drug users (76% versus 86%), and body mass index (22..8 versus 23.2 kg/m(2)). Nor did the nevirapine and lopinavir groups differ significantly in initial CD4 count, proportion with HCV genotype 1 or 4, use of PEG-IFN alfa-2a, weight-based RBV dosing, adherence to PEG-IFN/RBV, use of growth factor, or need for PEG-IFN or RBV dose reduction.

But the proportion of men was significantly lower in the nevirapine group (66% versus 81%, P = 0.03), fewer nevirapine takers had fibrosis stage F3 or greater (21% versus 52%, P = 0.001), fewer on nevirapine had cirrhosis (6% versus 32%, P = 0.001), initial HCV load was lower in the nevirapine group (median 5.7 versus 6.1 log IU/mL, P = 0.02), and a significantly lower proportion of people on nevirapine had a starting HCV load over 6 million IU/mL (44% versus 73%, P = 0.001).

In an intention-to-treat analysis, 40 people (56%) taking nevirapine achieved SVR with PEG-IFN/RBV, compared with 35 people (37%) taking lopinavir, a significant difference (P = 0.015). SVR rates were significantly better with nevirapine among people with genotype 1 or 4 (43% versus 25%, P = 0.04). A higher proportion with genotype 2 or 3 had an SVR with nevirapine than with lopinavir (78% versus 59%), though that difference stopped short of statistical significance (P = 0.1). Among people with an initial HCV load at or above 600,000 IU/mL, 58% taking nevirapine and 31% taking lopinavir had an SVR (P = 0.01) (from jules: the numbers of patients were small in this analysis). SVR rates did not differ significantly by liver fibrosis stage (F3-F4, 60% with nevirapine and 36% with lopinavir, P = 0.2).

Among reasons for failure to achieve SVR, nonresponse was the only one to differ in frequency between the nevirapine and lopinavir groups (8% versus 23%, P = 0.01). Rates of viral breakthrough were 7% with nevirapine and 14% with lopinavir (P = 0.1), and the relapse rate were higher with nevirapine (17% versus 11%, P = 0.2).

Multivariate analysis considering numerous factors that may influence SVR determined that three factors independently predicted SVR: a nevirapine regimen (P = 0.01), HCV genotype 2 or 3 (P < 0.001), and greater than 80% adherence to PEG-INF/RBV (P = 0.01). Taking nevirapine rather than lopinavir approximately doubled chances of achieving an SVR in this analysis.

The investigators proposed that lower HCV load with a nevirapine regimen may explain the better response to anti-HCV therapy in people taking a nevirapine-based antiretroviral regimen. They explained that HIV can induce HCV replication through TGF-beta1, which the liver produces in response to proinflammatory cytokines. Research has associated nevirapine with higher decreases in expression of TNF-alpha receptor than are achieved with other antiretrovirals [2], and the Spanish team proposed this might decrease TGF-beta1 secretion in the liver and thereby reduce HCV replication.

Session cochair and HIV/HCV expert Juergen Rockstroh suggested that in a study group of this size, multivariate analysis may not be able to correct for the distinct liver disease progression differences between the nevirapine group and the lopinavir group. He proposed that a matched-pair analysis may afford further insight into the relative value of nevirapine versus lopinavir/ritonavir in people starting PEG-IFN/RBV. Nevirapine must be used with caution in HCV-infected people because liver toxicity is among its best-documented side effects [3,4], especially in the first 10 weeks of therapy.

References

1. Mira JA, Lopez-Cortes LF, Vispo E, et al. Concomitant nevirapine therapy is associated with higher efficacy of pegylated interferon plus ribavirin among HIV/hepatitis C virus-coinfected patients. XVIII International AIDS Conference. July 18-23, 2010. Vienna. Abstract TUAB0101.

2. Virgili N, Fisac C, Martinez E, Ribera E, Gatell JM, Podzamczer D. Proinflammatory cytokine changes in clinically stable, virologically suppressed, HIV-infected patients switching from protease inhibitors to abacavir. J Acquir Immune Defic Syndr. 2009;50:552-553.

3. Macias J, Castellano V, Merchante N, et al. Effect of antiretroviral drugs on liver fibrosis in HIV-infected patients with chronic hepatitis C: harmful impact of nevirapine. AIDS. 2004;18:767-774.

4. Almond LM, Boffito M, Hoggard PG, et al. The relationship between nevirapine plasma concentrations and abnormal liver function tests. AIDS Res Hum Retroviruses. 2004;20:716-722.

Source