You are on page 1of 38

Accepted Manuscript

Title: Antiepileptic Drugs and Liver Disease

Author: Jorge Vidaurre, Satya Gedela, Shannon Yarosz

PII: S0887-8994(17)30637-9
DOI: https://doi.org/doi:10.1016/j.pediatrneurol.2017.09.013
Reference: PNU 9234

To appear in: Pediatric Neurology

Received date: 20-6-2017


Revised date: 5-9-2017
Accepted date: 19-9-2017

Please cite this article as: Jorge Vidaurre, Satya Gedela, Shannon Yarosz, Antiepileptic Drugs
and Liver Disease, Pediatric Neurology (2017),
https://doi.org/doi:10.1016/j.pediatrneurol.2017.09.013.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service
to our customers we are providing this early version of the manuscript. The manuscript will
undergo copyediting, typesetting, and review of the resulting proof before it is published in its
final form. Please note that during the production process errors may be discovered which could
affect the content, and all legal disclaimers that apply to the journal pertain.
Antiepileptic drugs and liver disease

Jorge Vidaurre, MD1, Satya Gedela, MD1, Shannon Yarosz, Pharm.D1

1
Division of Pediatric Neurology, Department of Pediatrics, Nationwide Children’s Hospital,

Columbus, Ohio

Running Title: Antiepileptic drugs and liver disease

Corresponding Author: Jorge Vidaurre, MD

700 Children’s Drive

Columbus, OH 43205

Tel: (614) 722-4730 Fax: (614) 722-4633

Jorge.Vidaurre@nationwidechildrens.org

Word count: Abstract, 229; Text: 4,292

Keywords: Antiepileptic drugs, hepatotoxicity, hepatic disease, seizures, epilepsy

The authors report no financial disclosures relevant to this work.

Page 1 of 37
Abstract

Acute, symptomatic seizures or epilepsy may complicate the course of hepatic disease.

Choosing the most appropriate antiepileptic drug (AED) in this setting represents a difficult

challenge, as most medications are metabolized by the liver. This article focuses on the acute and

chronic treatment of seizures in patients with advanced liver disease and reviews the hepatotoxic

potential of specific AEDs.

Newer AEDs with no, or minimal, hepatic metabolism, such as levetiracetam, lacosamide,

topiramate, gabapentin and pregabalin should be used as first line therapy. Medications

undergoing extensive hepatic metabolism, such as valproic acid, phenytoin and felbamate

should be used as drugs of last resort. In special circumstances, such as patients affected by

acute intermittent porphyria; exposure to most AEDs could precipitate attacks. In this clinical

scenario, bromides, levetiracetam, gabapentin and vigabatrin constitute safe choices. For the

treatment of status epilepticus, levetiracetam and lacosamide, available in IV preparations, are

good second line therapies after benzodiazepines fail to control seizures.

Hepatotoxicity is also a rare and unexpected side effect of AED therapy. Some drugs, such as

valproic acid, phenytoin and felbamate have a well-recognized association with liver toxicity.

Other AEDs, including phenobarbital, benzodiazepines, ethosuximide and the newer generations

of AEDs, have only rarely been linked to hepatotoxicity.

It is therefore necessary for physicians to be mindful of the pharmacokinetic profile and

hepatotoxic potential of the different AEDs available to treat patients affected by liver disease.

Introduction

Acute, symptomatic seizures or epilepsy may complicate the course of hepatic disease.

Page 2 of 37
Initiating antiepileptic therapy in this clinical scenario represents a difficult challenge, as most

medications are metabolized by the liver. Severe liver disease also affects the binding capacity of

AEDs to serum protein, increasing the risk of toxicity. Therefore, choosing the most appropriate

AED in this vulnerable population is extremely important.

Newer AEDs undergo no, or minimal, hepatic metabolism and constitute excellent therapeutic

choices. These medications have less potential for drug to drug interactions and lower protein

binding capacity. Some are available in IV formulations and can be used in emergency

situations. It is important for physicians to be familiar with these therapeutic choices and

understand the pharmacodynamics and pharmacokinetics of these drugs. This article focuses on

the acute and chronic treatment of seizures in patients with advance liver disease. The

hepatotoxic potential of different AEDs and special conditions which may require more specific

treatment, such as porphyria and Wilson disease is also reviewed.

Emergency treatment of seizures in patients with liver disease.

Patients affected by hepatic disease are at risk for recurrent seizures or status epilepticus. There

are no current standardized guidelines to assist physicians in this situation.

Multiple randomized control studies support the use of benzodiazepines as first line treatment

for status epilepticus in general.1 The RAMPART study showed a trend in favor of intramuscular

midazolam vs intravenous lorazepam for initial seizure control (73.4% vs 63.4%)2, likely

reflecting the speed of administration rather than the efficacy of the drug.

Benzodiazepines are metabolized predominantly in the liver and hepatic disease can alter their

metabolism. Midazolam is metabolized by the cytochrome P450 system and its clearance can be

affected by hepatic dysfunction. Lorazepam metabolism is minimally affected by liver disease3

and has fewer chances of drug interactions, making it a good choice for first line therapy (Table

Page 3 of 37
1). Benzodiazepines could potentially exacerbate any preexisting encephalopathy,4 but liver

injury is not a major concern.5 The choice of benzodiazepine should be based on availability of

the drug and presence or absence of an IV access (Figure 1).

If benzodiazepines fail to control seizures, second line agents are necessary. Commonly used

drugs for treatment of status epilepticus are valproic acid, phenytoin or fosphenytoin and

phenobarbital. Nevertheless, these are not ideal choices for patients with hepatic disease.

Valproic acid is mainly eliminated through the liver. Patients with hepatic disease could

potentially shift its metabolism to alternate pathways with the production of hepatotoxic

metabolites. 4, 6-9

Phenytoin has nonlinear kinetics and in the presence of hypoalbuminemia toxic levels can be

easily reached. 10-14

Phenobarbital is primarily metabolized in the liver and has a prolonged half-life of up to 126

hours5, which can be longer in the setting of liver disease. This could potentially complicate

encephalopathy in susceptible patients.15, 16

Newer AEDs such as levetiracetam and lacosamide constitute a more appropriate choice in this

group of patients. Both medications are available in IV formulations and have less chances of

exacerbating the hepatic injury. These drugs also have low or no binding to serum proteins1 with

fewer chances of drug to drug interactions, minimizing the risk of toxicity.

Levetiracetam has shown efficacy in the treatment of status epilepticus.17-22 The lack of major

side effects, such as hypotension or cardiac arrhythmias, makes it an ideal choice for critically ill

patients.21-23 Lacosamide, a newer AED with a favorable pharmacokinetic profile has also

showed efficacy in controlling status epilepticus and constitutes a good alternative.24-26

Page 4 of 37
If these drugs are not available or fail to control seizures, older AEDs can be used, but close

monitoring of drug levels is necessary. Phenobarbital can be an acceptable alternative, especially

if the risk of sedation is not a concern.

Long term AED therapy in patients with hepatic disease

Patients affected by liver disease may require long term AED treatment. The selection of AEDs

in this specific group deserves careful consideration.

Drug metabolism depends on the integrity of the hepatocyte, blood flow, and patency of

hepatobiliary system.5 Due to the large hepatic reserve, the liver dysfunction must be severe in

order to cause substantial alterations in drug metabolism.27 Mild to moderate liver disease may

need very minor dose adjustments, if any at all. Most AEDS are classified as “low extraction

drugs”, as they have an extraction of < 30% during the first passage through the liver. In clinical

practice, treatment can be started at regular doses but lower maintenance doses may be

required.28

Chronic management of seizures in patients with liver disease may become a difficult task.

Accurate calculation of drug adjustments is not possible, as there is no endogenous marker to

calculate hepatic clearance. Comorbidities such as renal failure and gastrointestinal dysmotility

affect the clearance or absorption of these drugs, resulting in toxicity or failure to control

seizures.29 Individual assessment of the severity of liver involvement is important and

consultation with a gastrointestinal specialist or hepatologist may be needed.

The newer generation of AEDs offers a more favorable pharmacokinetic profile than older

anticonvulsants and constitute a better option for long term treatment.

Page 5 of 37
In general, medications with minimal or no hepatic metabolism should be first line therapy and

drugs undergoing extensive hepatic metabolism should be avoided to the extent possible.

We provide a choice of different therapeutic options. Drugs are classified in different categories

from more to less suitable therapy, based on individual pharmacological characteristics and

chances of toxicity (Table 2).

First line therapy

Levetiracetam, a broad spectrum AED is an ideal first line therapy for patients with liver

disease.30 Sixty-six percent of the drug is excreted unchanged by the kidney, 24% undergoes

enzymatic hydrolysis and less than 2% is metabolized by hepatic enzymes.31 Levetiracetam has a

wide safety window with no major pharmacokinetic interactions. The dose needs no changes in

mild to moderate liver failure, but it should be decreased by half in patients with severe disease

and Child-Pugh class C (Table 3).31 Levetiracetam can effectively be utilized in patients on

venovenous hemofiltration.32

Lacosamide is metabolized by the CYP450, 2C19 system but has no major drug interactions and

the metabolites are inactive. Its protein binding capacity is less than 15% and 40% of the drug is

eliminated unchanged by the urine. Due to its linear pharmacokinetics, lacosamide constitutes as

a safe choice.15, 33

Topiramate is predominantly excreted by the kidneys as unmetabolized drug. Only 20% of the

dose is biotransformed by the liver. Topiramate is a potent anticonvulsant with a relatively low

potential for drug interactions, making it a reasonable choice for patients on multiple

medications. In severe liver disease, doses may need to be reduced by 30%.31, 34

Page 6 of 37
Gabapentin is mostly used for neuropathic pain, headaches and sleep disorders. Nevertheless,

due to his favorable pharmacokinetic profile, it’s another good alternative. It is excreted

unchanged in the urine and does not bind to serum proteins, minimizing the possibilities of drug

to drug interactions.35, 36

Pregabalin, similar to gabapentin, doesn’t bind to plasma proteins and is excreted by the

kidneys. In clinical practice, pregabalin is used mainly for treatment of neuropathic pain, but due

to its benign pharmacokinetics is a reasonable alternative in the treatment of seizures in patients

with hepatic disease.37, 38

Second line therapy

Vigabatrin is excreted unchanged by the kidneys without undergoing hepatic metabolism and is

not protein bound, so drug to drug interactions are not expected. Monitoring levels is not

necessary. Vigabatrin irreversibly affects gamma aminobutyric acid transaminase and the

duration of pharmacological effect is independent of serum concentration. 39 Vigabatrin is a very

good choice for patients with hepatic disease, but due to concerns of peripheral vision loss and

reversible MRI abnormalities,40, 41 it can be considered a second tier drug.

Oxcarbazepine may offer an advantage over carbamazepine in patients with liver disease. The

main metabolite is a monohydroxilated derivative, which is eliminated mainly by

glucoronidation.42-44 It is a weaker enzyme inducer with linear pharmacokinetics and less

potential for drug interactions. Oxcarbazepine also has fewer chances of hypersensitivity

reactions than carbamazepine.

Eslicarbazepine, a third generation member of the dibenzazepines, is similar to carbamazepine

and oxcarbazepine, but appears to have a more favorable profile than its predecessors.

Page 7 of 37
Eslicarbazepine is rapidly and extensively metabolized in the liver, but has minimal interactions

with cytochrome p450, minimizing the risk of drug to drug interactions.45-48

Zonisamide, a broad spectrum antiepileptic drug offers no significant hepatotoxicity. About

70% of the drug is cleared by the liver with 35% excreted unchanged by the kidneys. The

metabolism is through cytochrome P450 without production of active metabolites. Zonisamide

has linear pharmacokinetics and only 40% of the drug is protein bound, minimizing the risk of

drug interactions. Despite the favorable pharmacokinetic profile, the half-life can be up to 69

hours, so close monitoring is recommended.5, 49, 50

Third line therapy

Carbamazepine, an older generation sodium channel blocker, is mainly metabolized by the

liver. Carbamazepine undergoes auto induction and may produce transient elevation of hepatic

enzymes. Due to its enzyme inducing properties, there are higher chances of drug to drug

interactions than the newer generation sodium channel blockers.51-53

Perampanel, a recently approved glutamate receptor antagonist, undergoes extensive hepatic

metabolism by CYP3A4, but appears to have minimal effect on liver function. The half-life is

prolonged in patients with mild to moderate liver disease, so slow titration is recommended.54-58

Lamotrigine is primarily metabolized to the inactive N-2 nitrogen glucuronide metabolite by the

enzyme uridine diphosphate glucuronyl transferase and it should be used with caution in

moderate to severe liver disease. Recommendations are to reduce dose by 25% in moderate to

severe liver disease without ascites and 50% in patients with severe hepatic disease and ascites.28,
59

AEDs of last resort

Page 8 of 37
Valproic acid is not recommended in patients with liver dysfunction and should be avoided to

the extent possible. Despite the lack of strong evidence, valproic acid could potentially

exacerbate hepatic disease.5 Its binding to serum proteins is 90% and the unbound fraction

increases with chronic liver disease. This could lead to toxicity and increase the chances of drug

to drug interactions.

Phenytoin is metabolized by the cytochrome P450 and has been associated with hepatotoxicity.

Phenytoin has non-linear pharmacokinetics and frequent monitoring of blood levels is necessary

in order to avoid toxicity.60 . This is a special concern for patients with hypoalbuminemia.

Phenobarbital is metabolized by the liver, mostly by CTP2C9. It has a prolonged half-life and

could exacerbate a preexisting encephalopathy.61 Phenobarbital also has the potential for drug to

drug interactions. On the other hand, phenobarbital is a cheap medication, available worldwide

and has rarely been linked to hepatotoxicity. These characteristics could make it a reasonable

choice when newer AEDs are not available.

Felbamate is a medication used mainly in specialized centers for the treatment of patients with

Lennox Gastaut, Doose syndrome or refractory seizures.62 Felbamate is an enzyme inhibitor and

is metabolized by enzymes of the P450 system. It has potential for liver toxicity and significant

drug interactions.

Others

Tiagabine is not a widely used medication. There is no clear association with hepatotoxicity, but

is also metabolized by the liver by cytochrome P450 system and should not be a first line drug.63

Page 9 of 37
Ethosuximide is the drug of choice for absence seizures and can be used for specific epilepsy

syndromes. About 80% of the drug undergoes hepatic metabolism and 20% is excreted

unchanged. Close monitoring is advised when used in a patient with hepatic impairment.

Brivaracetam was recently approved by the Food and Drug Administration (FDA). This drug

was developed by modification on its predecessor levetiracetam. It has greater affinity towards

synaptic vesicle 2A (SV2A) and its binding capacity is low. Only 20% of the total in vivo

clearance is by the liver, so it is unlikely to be affected by significant drug interactions. Current

data is limited, but brivaracetam would likely be a safe medication for patients with liver disease.

Its clearance is reduced in patients with hepatic insufficiency and dose may need adjusting.64

In addition to individual pharmacokinetic properties of the different AEDs, other variables must

be considered when selecting therapy in patients affected by liver disease. These variables

include formulations and cost of medications, associated comorbid conditions and seizure type.

First or second line agents are effective in treating multiple seizure types.

For generalized convulsive seizures, including myoclonus and generalized tonic - clonic

seizures, broad spectrum drugs, such as levetiracetam , topiramate or zonisamide are good

choices.65-67

For absence seizures in children, ethosuximide is the drug of choice.68 Due to favorable

pharmacokinetics, levetiracetam can be considered first.69 There is also data from small studies

about efficacy of zonisamide and topiramate.70

For seizures of focal onset levetiracetam continues to be an ideal choice, but virtually any first or

second line therapy can be considered, including sodium channel blockers such as lacosamide,

oxcarbazepine and eslicarbazepine71, 72

Page 10 of 37
Hepatotoxicity caused by AEDs.

Chemicals that cause liver injury are call hepatotoxins. There are more than 900 drugs, toxins

and herbs that can produce liver damage.73

In order to determine the hepatotoxic potential of a specific drug, a high number of patients need

to be exposed to the drug and the risk for hepatotoxicity is usually discovered in the

postmarketing phase.13

Drug induced liver injury (DILI) is a rare but potential side effect of AEDs. Serious liver injury

may be the cause of disapproval or removal from the market of a new compound. In patients

receiving liver transplantation in the USA between 1990 and 2002, AEDs were the third most

common cause.74

AEDs can commonly cause asymptomatic or transient elevations in the liver function test.

Elevation in gamma-glutamyl transferase is observed in up to 75-95% of patients exposed to

enzyme inducers, such as carbamazepine, and phenobarbital.53

Zimmerman in 1960 showed that an increase in alanine transferase ≥3 the upper limit of normal

(x ULN) and total bilirubin level ≥2 × ULN (especially in the presence of jaundice) may lead to a

mortality rate of 10-50%.75

In 2011, the new consensus criteria defined DILI as:

(1) Increase in alanine transferase ≥3 x ULN and total bilirubin levels ≥2 × ULN;

(2) increase in alanine transferase level ≥5 × ULN;

Page 11 of 37
(3) increase in alkaline phosphatase level ≥2 × ULN (especially when associated to elevations in

γ-glutamyl transpeptidase).76

These values are useful in clinical practice, as it’s difficult to predict the occurrence of DILI,

before the clinical course becomes irreversible. The mechanism of liver injury caused by AEDS

is heterogeneous. Hypersensitivity features are found in 70% of cases of DILI related to

phenytoin, but only 30% of cases associated with carbamazepine. Hypersensitivity is usually not

observed in valproic acid. The hepatotoxic potential between different AEDs is also variable and

a detailed analysis is presented in this article.

AEDs with well recognized hepatotoxicity

Valproic Acid

The hepatotoxic potential of valproic acid is well recognized. Valproic acid is the third most

common cause of drug induced liver fatalities reported by the world health organization.77 The

mechanism of injury is unclear, but it may be related to interference with mitochondrial beta

oxidation of fatty acids.78 An inherited or acquired impairment in beta -oxidation of valproate

may contribute to the liver toxicity.79 Hepatic injury can be of different types, the most common

type being asymptomatic elevation of liver function tests, which does not require discontinuation

of the drug.80 More severe reactions are rare, occurring in about 1 per 15,000 exposures.7, 81

These severe reactions are usually observed in the first three months of treatment.82, 83 In cases of

significant elevation of hepatic enzymes of more than 3 x ULN, valproic acid should be

discontinued.

Clinical symptoms of hepatotoxicity are nausea, vomiting, increase in seizures, jaundice and

fatigue, but signs of hypersensitivity are usually lacking.73 There are clear risk factors identified

in valproic acid associated hepatotoxicity. These include younger age

Page 12 of 37
and polytherapy (Table 4). Additional risk factors include developmental delay, metabolic

disorders, febrile illness and status epilepticus.84, 85

Patients bearing mutations within the mitochondrial DNA replicase, polymerase gamma encoded

by the POLG gene, responsible for the Alpers-Huttenlocher syndrome are at high risk of liver

failure from valproic acid exposure. Testing for POLG mutations may be necessary in children

with refractory seizures and developmental regression before use of valproic acid.86, 87 Despite

available testing, the diagnosis of POLG related disorders remains challenging. The degree of

involvement can be milder and there is a high degree of phenotypic variation.88-91

Valproic acid has also been associated with Reye-like syndrome, which manifests with lethargy,

fever, vomiting and microvesicular steatosis on liver biopsy.92

Asymptomatic hyperammonemia is a common abnormality observed during valproate therapy

and it doesn’t require discontinuation of the drug.93, 94 Valproic acid hyperamonemic

encephalopathy is a more serious condition. Symptoms consist of progressive confusional state

which can lead to coma and death.95, 96 Hepatic enzymes may be normal, but glutamine in CSF

can be high.97 Concomitant use of AEDS, especially topiramate can increase risk of developing

this complication.98

Phenytoin

Phenytoin is an aromatic anticonvulsant with enzyme inducing properties. Its association with

hepatotoxicity is also well recognized.77 Phenytoin produces transient and asymptomatic increase

in ALT in 25% of patients.99 Severe liver injury is part of an idiosyncratic reaction, occurring in

genetically predisposed patients.100 Hepatotoxicity occurs with a frequency of 1 per 10,000 to 1

per 50,000 exposures.101 The incidence is higher in blacks. Children appear to be less

Page 13 of 37
susceptible.102 Liver toxicity usually occurs during the first six weeks of treatment and is likely

related to the production of arene oxide and cathecol metabolites in liver microsomes.103 These

metabolites may trigger an oxidative injury with a secondary immune mediated response. The

majority of cases are associated with hypersensitivity reactions.104 Fever is observed in 75% of

cases, rash in 62% and eosinophilia in 89%.102 Pathological studies show variable types of

injury, including granuloma formation, cholestatic, cytotoxic or mixed pattern. The cytotoxic

pattern is the most common and the mortality rate after severe liver involvement is 13%.102, 103

Carbamazepine

Carbamazepine is another aromatic drug which is metabolized by epoxidation and hydroxylation

and is a well-established cause of hepatotoxicity. Carbamazepine induces its own metabolism

and produces a transient, asymptomatic elevation in hepatic enzymes in 10% of patients exposed

to the drug.105 Severe hepatotoxicity is rare, unpredictable, not dose dependent and can occur in

the first 12 months of therapy. The likelihood of hepatic death appears higher in children,

especially when exposed to multiple AEDs.106

Carbamazepine can also produce intermediate arene oxides, but unlike phenytoin, only 30-40%

of cases are linked to hypersensitivity reactions. Once hepatotoxicity has developed, the

mortality rate is about 25%.60 Liver biopsies show inflammatory reactions, cholestasis or

hepatocellular injury with hepatic necrosis observed in lethal cases.103 Other types of liver injury

include granulomatous hepatitis and ductopenia, a rare condition with loss of small bile ducts

and jaundice, also associated with valproic acid and lamotrigine exposure.107, 108

Felbamate

Page 14 of 37
Felbamate was synthetized in 1950, while exploring anxiolytic substances. Its antiepileptic

properties were discovered 30 years later and approved by the FDA in 1993. Felbamate was the

first new AED after the approval of valproic acid in 1978. This drug was widely used until the

reports of aplastic anemia and liver failure in the post marketing phase. Subsequent analysis

showed that the risk of liver failure is between 1:26,000 to 1:34,000 (which may be lower than

the risk of valproic acid associated hepatotoxicity). The risk factors associated with hepatic

failure are less defined. Female gender and polytherapy could increase this risk.109 The mean

time of presentation is between 6- 12 months of therapy. Liver failure can occur without warning

and be fulminant. Patients should be advised about recognition of clinical symptoms. Felbamate

should be discontinued with aspartate aminotransferase – alanine transferase elevations of ≥2 ×

ULN.110

AEDS with low or unclear hepatotoxic potential

Oxcarbazepine

Oxcarbazepine is a newer generation sodium channel blocker. Unlike carbamazepine its older

relative, the association with hepatotoxicity is rare. Most cases occurred with previous exposure

to another AED or in the setting of elevated liver enzymes. There are isolated reports of

anticonvulsant hypersensitivity syndrome and hepatitis.111-113

Topiramate

Topiramate can produce hyperamonemia when combined with valproic, but hepatotoxicity when

used in monotherapy is very rare. There are few reports of severe hepatotoxicity including one

case leading to liver transplant in a patient also receiving carbamazepine.114, 115

Phenobarbital

Page 15 of 37
Phenobarbital is one of the oldest aromatic AEDs, available since 1911. Despite its extensive

use, phenobarbital has only been associated to hepatotoxicity in rare instances. Phenobarbital can

produce elevation in liver enzymes, and it has been occasionally linked to hypersensitivity

reactions, hepatitis and acute liver failure.116-118 Oxidative stress in the hepatic mitochondria may

be one of the mechanisms of hepatic injury.119

Lamotrigine

Lamotrigine is an effective and relatively safe medication. Rare cases of severe hepatotoxicity

have been reported. The liver dysfunction can be reversible upon drug discontinuation.

Lamotrigine is an aromatic drug and can produce liver toxicity as part of a general

hypersensitivity reaction. Patients with comorbid conditions, such as status epilepticus and those

receiving polytherapy (especially valproate and carbamazepine) are more susceptible to

hepatotoxicity.120-122

Levetiracetam

Levetiracetam does not interact with the cytochrome P450 system and is the drug of first choice

for patients with liver disease. Nevertheless, there are rare reports of hepatotoxicity, ranging

from elevations of liver enzymes to severe hepatotoxicity or liver failure. The physiopathology

of liver toxicity is unknown.123-127

Lacosamide

Lacosamide is a new generation AED, which is 40% excreted unchanged by the urine. Part of the

drug is metabolized by the liver and there are only rare reports of hepatic enzymes elevation and

liver toxicity.128-130

Page 16 of 37
Zonisamide

Zonisamide, a second generation medication has been rarely linked to hepatotoxicity. There are a

few reports of liver toxicity, and “vanishing bile duct syndrome”. Patients with chronic cirrhosis

may be at risk.131, 132

Ethosuximide

Evidence linking Ethosuximide to hepatotoxity is lacking with exception of few reports, mainly

related to its use in combination with other drugs, usually valproic acid .133, 134

Benzodiazepines

Despite the wide use of benzodiazepines, an association with liver toxicity is not strong and

extremely rare.135-137

Gabapentin

Gabapentin has no appreciably liver metabolism and there are only rare individual case reports

about possible hepatotoxicity. Some of these cases include patients taking multiple drugs and the

association remains unclear.138-141

Pregabalin

Pregabalin does not inhibit or induce liver enzymes. Despite its favorable pharmacokinetics,

there are few reports about potential liver toxicity, with onset in the first 2 weeks of treatment.

Cholestatic and hepatocellular patterns of injury have been described.142-145

Vigabatrin

Page 17 of 37
Vigabatrin is a drug which lacks hepatic metabolism and hepatotoxicity has rarely been

reported.146, 147

Eslicarbazepine

Eslicarbazepine is a drug related to oxcarbazepine and there are only isolated reports about liver

injury , usually with rapid recovery after drug discontinuation.148

AEDs with no known association with hepatotoxicity

Drugs such as tiagabine, perampanel and brevaracetam have not yet been linked to liver

toxicity.58

Despite the available information, most cases of hepatoxicity associated with AEDs are

unexpected and difficult to predict. An increase in liver function tests of 2-3 folds from baseline

warrant enhanced vigilance. Once laboratory values surpass this range or the diagnosis of DILI

has been made, prompt discontinuation of the drug should follow. Other specific treatment

modalities have shown some benefit (Table 5).122, 147, 149-156 Liver transplantation is the treatment

of choice for end stage hepatic damage

AED therapy in special circumstances

Acute porphyria

The acute porphyrias are a group of rare inherited disorders of haem synthesis. Acute

intermittent porphyria is the most common of the hepatic porphyrias and is caused by a

deficiency of hydroxymethylbilane synthase involved in heme synthesis. Most affected

individuals remain asymptomatic, but acute attacks can suddenly be precipitated by exposure to

multiple drugs. Symptoms usually consist of abdominal pain, symmetric motor neuropathy,

Page 18 of 37
confusion and seizures. During a crisis there is elevation of the porphyrins precursors δ-

aminolevulinic acid (ALA) and porphobilinogen (PBG).157

The prevalence of seizures in patients with acute intermittent porphyria is about 3.7%, but this

number can increase to. 5.1 % in patients who already presented with acute attacks.158 Selecting

the most appropriate AED remains problematic, as most anticonvulsants are porphorygenic.

Enzyme inducers, such as carbamazepine, phenytoin and phenobarbital can precipitate attacks,

likely by an increase in catabolism of hydroxymethylbilane synthetase and uroporphyrinogen

decarboxylase, affecting haem synthesis.159, 160

Other AEDS including lamotrigine, valproic, primidone, tiagabine, ethosuximide, topiramate,

methsuximide, trimethadione and felbamate have been associated with clinical worsening or

demonstrated porphyrin accumulation in chick embryo liver cells.161-168

Bromides have been historically the drug of choice until new AEDs with more favorable kinetics

appeared in the 1990s (Table 6).169-172 AEDs such as levetiracetam and gabapentin with

negligible hepatic metabolism should be first line treatment. Vigabatrin lacks hepatic metabolism

and constitutes a safe option, but has the potential of causing visual field defects.159, 167

Oxcarbazepine with low hepatic induction of microsomal enzymes has been tried successfully.173

Carbamazepine, a drug with porphorygenic potential has been used in isolated cases and it can be

an alternative when no other options are available, especially in patients with normal ALA and

PBG levels.174

For acute treatment, benzodiazepines are good therapeutic choices. Clonazepam have been tried

safely, but at higher doses could exacerbate symptoms.160, 161, 175 Levetiracetam is available in IV

preparations and is another good alternative for emergency situations.

If general anesthesia is needed midazolam and propofol are probably safe medications.176

Page 19 of 37
Wilson’s disease

Wilson’s disease is a rare autosomal disorder of altered copper transport by loss of function of

the ATP7B copper – binding protein, resulting in accumulation of copper in multiple organs.

Laboratory testing shows low ceruloplasmin and increase in 24 hour urine copper assay.177

Hepatic and neurological involvement are prominent features. Hepatic involvement can vary

from elevation of liver enzymes to hepatic failure or cirrhosis

Neurological symptoms consist of movement disorder and cognitive deterioration, but seizures

can be observed in about 5.5 % of patients before initiation of therapy and in 2.8 % during de-

coppering treatment.178 Seizures can be focal or generalized and can present as status epilepticus.

Good seizure control is achieved in most patients and only a small proportion may become

refractory.178, 179 Multiple AEDS, including phenytoin, valproic acid , benzodiazepines and

carbamazepine have been used successfully; however newer AEDs with less hepatic metabolism

should be favored, especially if the hepatic function is severely compromised. It is important to

supplement with pyridoxine during penicillamine therapy, as pyridoxine deficiency can cause

seizures during the chelation treatment.180

Conclusion

Hepatic disease and seizures may coexist and the use of anticonvulsant therapy is indicated.

Selecting the most appropriate AED therapy remains challenging. The goal of therapy is to

maximize efficacy and minimize toxicity. The ideal AED should have good efficacy, minimal or

no hepatic metabolism and no significant binding to serum proteins.

Newer AEDs with a more favorable pharmacokinetic profile and with lower risk of toxicity offer

good therapeutic choices for this subset of patients. . These medications are also ideal first line

Page 20 of 37
treatment for patients affected by rare conditions, like porphyria or Wilson’s disease with severe

hepatic involvement. Some of these drugs, such as levetiracetam and lacosamide are available in

IV preparations and can be used in emergency situations. Hepatotoxicity is also a rare but

potential side effect of AEDs and identification of high risk populations is important to prevent

liver injury.

It is therefore necessary for clinicians to be aware of the different therapeutic options available

for the treatment of seizures in patients with liver disease and recognize the hepatotoxic potential

of the different AEDs.

Page 21 of 37
References

1. Alvarez V, Rossetti AO. Monotherapy or polytherapy for first-line treatment of SE? J Clin
Neurophysiol 2016;33:14-17.
2. Silbergleit R, Durkalski V, Lowenstein D, et al. Intramuscular versus intravenous therapy for
prehospital status epilepticus. N Engl J Med 2012;366:591-600.
3. Greenblatt DJ. Clinical pharmacokinetics of oxazepam and lorazepam. Clinical Pharmacokinet
1981;6:89-105.
4. Ruiz-Gimenez J, Sanchez-Alvarez JC, Canadillas-Hidalgo F, Serrano-Castro PJ. Antiepileptic
treatment in patients with epilepsy and other comorbidities. Seizure 2010;19:375-382.
5. Ahmed SN, Siddiqi ZA. Antiepileptic drugs and liver disease. Seizure 2006;15:156-164.
6. Sztajnkrycer MD. Valproic acid toxicity: overview and management. J Toxicol Clin Toxicol
2002;40:789-801.
7. Dreifuss FE, Langer DH, Moline KA, Maxwell JE. Valproic acid hepatic fatalities. II. US
experience since 1984. Neurology 1989;39:201-207.
8. Willmore LJ, Wilder BJ, Bruni J, Villarreal HJ. Effect of valproic acid on hepatic function.
Neurology 1978;28:961-964.
9. Neuman MG, Nanau RM, Shekh-Ahmad T, Yagen B, Bialer M. Valproic acid derivatives signal
for apoptosis and repair in vitro. Clinical Biochem 2013;46:1532-1537.
10. Bajpai M, Roskos LK, Shen DD, Levy RH. Roles of cytochrome P4502C9 and cytochrome
P4502C19 in the stereoselective metabolism of phenytoin to its major metabolite. Drug Metab Dispos
1996;24:1401-1403.
11. Glazko AJ, Chang T, Baukema J, Dill WA, Goulet JR, Buchanan RA. Metabolic disposition of
diphenylhydantoin in normal human subjects following intravenous administration. Clin Pharmacol Ther
1969;10:498-504.
12. Sasaki E, Iwamura A, Tsuneyama K, et al. Role of cytochrome P450-mediated metabolism and
identification of novel thiol-conjugated metabolites in mice with phenytoin-induced liver injury. Toxicol
Lett 2015;232:79-88.
13. Bjornsson E. Hepatotoxicity associated with antiepileptic drugs. Acta Neurol Scand
2008;118:281-290.
14. Mendis GP, Gibberd FB, Hunt HA. Plasma activities of hepatic enzymes in patients on
anticonvulsant therapy. Seizure 1993;2:319-323.
15. Romigi A, Placidi F, Liguori C, et al. Lacosamide as add-on treatment of focal symptomatic
epilepsy in a patient with alcoholic liver cirrhosis. Epilepsy Behav 2014;2:161-163.
16. Alvin J, McHorse T, Hoyumpa A, Bush MT, Schenker S. The effect of liver disease in man on
the disposition of phenobarbital. J Pharmacol Exp Ther 1975;192:224-235.
17. Misra UK, Kalita J, Maurya PK. Levetiracetam versus lorazepam in status epilepticus: a
randomized, open labeled pilot study. J Neurol 2012;259:645-648.
18. Zheng F, Du C, Wang X. Levetiracetam for the treatment of status epilepticus. Exp Rev
Neurother 2015;15:1113-1121.
19. Mundlamuri RC, Sinha S, Subbakrishna DK, et al. Management of generalised convulsive status
epilepticus (SE): A prospective randomised controlled study of combined treatment with intravenous
lorazepam with either phenytoin, sodium valproate or levetiracetam--Pilot study. Epilepsy Res
2015;114:52-58.
20. Yasiry Z, Shorvon SD. The relative effectiveness of five antiepileptic drugs in treatment of
benzodiazepine-resistant convulsive status epilepticus: a meta-analysis of published studies. Seizure
2014;23:167-174.
21. Nau KM, Divertie GD, Valentino AK, Freeman WD. Safety and efficacy of levetiracetam for
critically ill patients with seizures. Neurocrit Care 2009;11:34-37.
22. Shin HW, Davis R. Review of Levetiracetam as a first line treatment in status epilepticus in the
adult patients - What do we know so far? Front Neurol 2013;4:111.

Page 22 of 37
23. Lyseng-Williamson KA. Spotlight on levetiracetam in epilepsy. CNS Drugs 2011;25:901-905.
24. Legros B, Depondt C, Levy-Nogueira M, et al. Intravenous lacosamide in refractory seizure
clusters and status epilepticus: comparison of 200 and 400 mg loading doses. Neurocrit Care
2014;20:484-488.
25. Grosso S, Zamponi N, Bartocci A, et al. Lacosamide in children with refractory status epilepticus.
A multicenter Italian experience. Eur J Pediatr Neurol 2014;18:604-608.
26. Hofler J, Unterberger I, Dobesberger J, Kuchukhidze G, Walser G, Trinka E. Intravenous
lacosamide in status epilepticus and seizure clusters. Epilepsia 2011;52:e148-152.
27. Asconape JJ, Penry JK. Use of antiepileptic drugs in the presence of liver and kidney diseases: a
review. Epilepsia 1982;23 Suppl 1:S65-79.
28. Delco F, Tchambaz L, Schlienger R, Drewe J, Krahenbuhl S. Dose adjustment in patients with
liver disease. Drug Saf 2005;28:529-545.
29. Borges de Lacerda GC. Treating seizures in renal and hepatic failure. J Epilepsy Clin
Neurophysiol 2008;14:46-50.
30. Lin CH, Chen CL, Lin TK, Chen NC, Tsai MH, Chuang YC. Levetiracetam in the treatment of
epileptic seizures after liver transplantation. Medicine (Baltimore) 2015;94:e1350.
31. Brockmoller J, Thomsen T, Wittstock M, Coupez R, Lochs H, Roots I. Pharmacokinetics of
levetiracetam in patients with moderate to severe liver cirrhosis (Child-Pugh classes A, B, and C):
characterization by dynamic liver function tests. Clin Pharmacol Ther 2005;77:529-541.
32. New AM, Nei SD, Kashani KB, Rabinstein AA, Frazee EN. Levetiracetam pharmacokinetics
during continuous venovenous hemofiltration and acute liver dysfunction. Neurocrit Care 2016;25:141-
144.
33. Asconape JJ. Use of antiepileptic drugs in hepatic and renal disease. Handb Clin Neurol
2014;119:417-432.
34. Patsalos PN. Pharmacokinetic profile of levetiracetam: toward ideal characteristics. Pharmacol
Ther 2000;85:77-85.
35. Calandre EP, Rico-Villademoros F, Slim M. Alpha2delta ligands, gabapentin, PGB and MGB: a
review of their clinical pharmacology and therapeutic use. Exp Rev Neurother 2016:1-15.
36. Vollmer KO, von Hodenberg A, Kolle EU. Pharmacokinetics and metabolism of gabapentin in
rat, dog and man. Arzneimittelforschung 1986;36:830-839.
37. Offord J, Isom LL. Drugging the undruggable: gabapentin, pregabalin and the calcium channel
alpha2delta subunit. Crit Rev Biochem Mol Biol 2015;51:246-256.
38. Bockbrader HN, Wesche D, Miller R, Chapel S, Janiczek N, Burger P. A comparison of the
pharmacokinetics and pharmacodynamics of pregabalin and gabapentin. Clin Pharmacokinet
2010;49:661-669.
39. Schechter PJ. Clinical pharmacology of vigabatrin. Br J Clin Pharmacol 1989;27 Suppl 1:19S-
22S.
40. Westall CA, Wright T, Cortese F, Kumarappah A, Snead OC, 3rd, Buncic JR. Vigabatrin retinal
toxicity in children with infantile spasms: An observational cohort study. Neurology 2014;83:2262-2268.
41. Rasmussen AD, Richmond E, Wegener KM, Downes N, Mullins P. Vigabatrin-induced CNS
changes in juvenile rats: Induction, progression and recovery of myelin-related changes. Neurotoxicol
2015;46:137-144.
42. Lloyd P, Flesch G, Dieterle W. Clinical pharmacology and pharmacokinetics of oxcarbazepine.
Epilepsia 1994;35 Suppl 3:S10-13.
43. Flesch G. Overview of the clinical pharmacokinetics of oxcarbazepine. Clin Drug Invest
2004;24:185-203.
44. Reinikainen KJ, Keranen T, Halonen T, Komulainen H, Riekkinen PJ. Comparison of
oxcarbazepine and carbamazepine: a double-blind study. Epilepsy Res 1987;1:284-289.
45. Shorvon SD, Trinka E, Steinhoff BJ, et al. Eslicarbazepine acetate: its effectiveness as adjunctive
therapy in clinical trials and open studies. J Neurol 2017;264:421-431.

Page 23 of 37
46. Almeida L, Potgieter JH, Maia J, Potgieter MA, Mota F, Soares-da-Silva P. Pharmacokinetics of
eslicarbazepine acetate in patients with moderate hepatic impairment. Eur J Clin Pharmacol 2008;64:267-
273.
47. Ben-Menachem E. Eslicarbazepine acetate: a well-kept secret? Epilepsy Curr 2010;10:7-8.
48. Bialer M, Soares-da-Silva P. Pharmacokinetics and drug interactions of eslicarbazepine acetate.
Epilepsia 2012;53:935-946.
49. Wilfong AA, Willmore LJ. Zonisamide - a review of experience and use in partial seizures.
Neuropsychiatr Dis Treat 2006;2:269-280.
50. Jacob S, Nair AB. An Updated Overview on Therapeutic Drug Monitoring of Recent
Antiepileptic Drugs. Drugs R D 2016;16:303-316.
51. Pearce RE, Lu W, Wang Y, Uetrecht JP, Correia MA, Leeder JS. Pathways of carbamazepine
bioactivation in vitro. III. The role of human cytochrome P450 enzymes in the formation of 2,3-
dihydroxycarbamazepine. Drug Metab Dispos 2008;36:1637-1649.
52. Bertilsson L, Tomson T. Clinical pharmacokinetics and pharmacological effects of
carbamazepine and carbamazepine-10,11-epoxide. An update. Clin Pharmacokinet 1986;11:177-198.
53. Strolin Benedetti M, Ruty B, Baltes E. Induction of endogenous pathways by antiepileptics and
clinical implications. Fundam Clin Pharmacol 2005;19:511-529.
54. Patsalos PN. The clinical pharmacology profile of the new antiepileptic drug perampanel: A
novel noncompetitive AMPA receptor antagonist. Epilepsia 2015;56:12-27.
55. Rogawski MA, Hanada T. Preclinical pharmacology of perampanel, a selective non-competitive
AMPA receptor antagonist. Acta Neurol Scand Supp 2013:19-24.
56. Strzelczyk A, Willems LM, Willig S, Rosenow F, Bauer S. Perampanel in the treatment of focal
and idiopathic generalized epilepsies and of status epilepticus. Exp Rev Clin Pharmacol 2015;8:733-740.
57. Rektor I, Krauss GL, Bar M, et al. Perampanel Study 207: long-term open-label evaluation in
patients with epilepsy. Acta Neurol Scand 2012;126:263-269.
58. Laurenza A, Yang H, Williams B, Zhou S, Ferry J. Absence of Liver Toxicity in Perampanel-
Treated Subjects: Pooled results from partial seizure phase III perampanel clinical studies. Epilepsy Res
2015;113:76-85.
59. Hurley SC. Lamotrigine update and its use in mood disorders. Ann Pharmacother 2002;36:860-
873.
60. Dreifuss FE, Langer DH. Hepatic considerations in the use of antiepileptic drugs. Epilepsia
1987;28 Suppl 2:S23-29.
61. Pacifici GM. Clinical pharmacology of phenobarbital in neonates: effects, metabolism and
pharmacokinetics. Curr Pediatr Rev 2016;12:48-54.
62. Burdette DE, Sackellares JC. Felbamate pharmacology and use in epilepsy. Clin Neuropharmacol
1994;17:389-402.
63. Brodie MJ. Tiagabine pharmacology in profile. Epilepsia 1995;36 Suppl 6:S7-S9.
64. von Rosenstiel P. Brivaracetam (UCB 34714). Neurotherapeutics 2007;4:84-87.
65. Beydoun A, D'Souza J. Treatment of idiopathic generalized epilepsy - a review of the evidence.
Expert Opin Pharmacother 2012;13:1283-1298.
66. Coppola G, Piccorossi A, Operto FF, Verrotti A. Anticonvulsant drugs for generalized tonic-
clonic epilepsy. Expert Opin Pharmacother 2017;18:925-936.
67. Mantoan L, Walker M. Treatment options in juvenile myoclonic epilepsy. Curr Treat Options
Neurol 2011;13:355-370.
68. Glauser TA, Cnaan A, Shinnar S, et al. Ethosuximide, valproic acid, and lamotrigine in childhood
absence epilepsy: initial monotherapy outcomes at 12 months. Epilepsia 2013;54:141-155.
69. Hughes JR. Absence seizures: a review of recent reports with new concepts. Epilepsy Behav
2009;15:404-412.
70. Velizarova R, Crespel A, Genton P, Serafini A, Gelisse P. Zonisamide for refractory juvenile
absence epilepsy. Epilepsy Res 2014;108:1263-1266.

Page 24 of 37
71. Bauer S, Willems LM, Paule E, et al. The efficacy of lacosamide as monotherapy and adjunctive
therapy in focal epilepsy and its use in status epilepticus: clinical trial evidence and experience. Ther Adv
Neurol Disord 2017;10:103-126.
72. Coppola G, Iapadre G, Operto FF, Verrotti A. New developments in the management of partial-
onset epilepsy: role of brivaracetam. Drug Des Devel Ther 2017;11:643-657.
73. Pandit A, Sachdeva T, Bafna P. Drug-induced hepatotoxicity: a review. J Appl Pharm Sci
2012;2:233-243.
74. Russo MW, Galanko JA, Shrestha R, Fried MW, Watkins P. Liver transplantation for acute liver
failure from drug induced liver injury in the United States. Liver Transpl 2004;10:1018-1023.
75. Bjornsson E, Olsson R. Outcome and prognostic markers in severe drug-induced liver disease.
Hepatology 2005;42:481-489.
76. Robles-Diaz M, Lucena MI, Kaplowitz N, et al. Use of Hy's law and a new composite algorithm
to predict acute liver failure in patients with drug-induced liver injury. Gastroenterology 2014;147:109-
118 e105.
77. Bjornsson E, Olsson R. Suspected drug-induced liver fatalities reported to the WHO database.
Dig Liver Dis 2006;38:33-38.
78. Coulter DL. Carnitine, valproate, and toxicity. J Child Neurol 1991;6:7-14.
79. Siemes H, Nau H, Schultze K, et al. Valproate (VPA) metabolites in various clinical conditions of
probable VPA-associated hepatotoxicity. Epilepsia 1993;34:332-346.
80. Powell-Jackson PR, Tredger JM, Williams R. Hepatotoxicity to sodium valproate: a review. Gut
1984;25:673-681.
81. Koenig SA, Buesing D, Longin E, et al. Valproic acid-induced hepatopathy: nine new fatalities in
Germany from 1994 to 2003. Epilepsia 2006;47:2027-2031.
82. Suchy FJ, Balistreri WF, Buchino JJ, et al. Acute hepatic failure associated with the use of
sodium valproate. N Engl J Med 1979;300:962-966.
83. Caparros-Lefebvre D, Lecomte-Houcke M, Pruvot FR, Declerck N, Paris JC, Petit H. Unusual
electronmicroscopic changes in valproate-associated liver failure. Lancet 1993;341:1604.
84. Dreifuss FE, Santilli N, Langer DH, Sweeney KP, Moline KA, Menander KB. Valproic acid
hepatic fatalities: a retrospective review. Neurology 1987;37:379-385.
85. Bryant AE, 3rd, Dreifuss FE. Valproic acid hepatic fatalities. III. U.S. experience since 1986.
Neurology 1996;46:465-469.
86. Stewart JD, Horvath R, Baruffini E, et al. Polymerase gamma gene POLG determines the risk of
sodium valproate-induced liver toxicity. Hepatology 2010;52:1791-1796.
87. Rust RS. Alpers-huttenlocher syndrome: origins of clinicopathologic recognition. Pediatr Neurol
2013;48:165-166.
88. Stumpf JD, Saneto RP, Copeland WC. Clinical and molecular features of POLG-related
mitochondrial disease. Cold Spring Harb Perspect Biol 2013;5:a011395.
89. Burusnukul P, de los Reyes EC. Phenotypic variations in 3 children with POLG1 mutations. J
Child Neurol 2009;24:482-486.
90. Rajakulendran S, Pitceathly RD, Taanman JW, et al. A Clinical, Neuropathological and Genetic
Study of Homozygous A467T POLG-Related Mitochondrial Disease. Plos One 2016;11:e0145500.
91. Da Pozzo P, Cardaioli E, Rubegni A, et al. Novel POLG mutations and variable clinical
phenotypes in 13 Italian patients. Neurol Sci 2017;38:563-570.
92. Gerber N, Dickinson RG, Harland RC, et al. Reye-like syndrome associated with valproic acid
therapy. J Pediatr 1979;95:142-144.
93. Murphy JV, Marquardt K. Asymptomatic hyperammonemia in patients receiving valproic acid.
Arch Neurol 1982;39:591-592.
94. Nicolai J, Carr RB. The measurement of ammonia blood levels in patients taking valproic acid:
looking for problems where they do not exist? Epilepsy Behav 2008;12:494-496; discussion 497-498.
95. Gerstner T, Buesing D, Longin E, et al. Valproic acid induced encephalopathy--19 new cases in
Germany from 1994 to 2003--a side effect associated to VPA-therapy not only in young children. Seizure
2006;15:443-448.

Page 25 of 37
96. Kifune A, Kubota F, Shibata N, Akata T, Kikuchi S. Valproic acid-induced hyperammonemic
encephalopathy with triphasic waves. Epilepsia 2000;41:909-912.
97. Vossler DG, Wilensky AJ, Cawthon DF, et al. Serum and CSF glutamine levels in valproate-
related hyperammonemic encephalopathy. Epilepsia 2002;43:154-159.
98. Blackford MG, Do ST, Enlow TC, Reed MD. Valproic Acid and topiramate induced
hyperammonemic encephalopathy in a patient with normal serum carnitine. J Pediatr Pharmacol Ther
2013;18:128-136.
99. Wall M, Baird-Lambert J, Buchanan N, Farrell G. Liver function tests in persons receiving
anticonvulsant medications. Seizure 1992;1:187-190.
100. Spielberg SP, Gordon GB, Blake DA, Goldstein DA, Herlong HF. Predisposition to phenytoin
hepatotoxicity assessed in vitro. N Engl J Med 1981;305:722-727.
101. Farrell GC. Drug-induced liver disease. Edinburgh ; New York: Churchill Livingstone, 1994.
102. Mullick FG, Ishak KG. Hepatic injury associated with diphenylhydantoin therapy. A
clinicopathologic study of 20 cases. Am J Clin Pathol 1980;74:442-452.
103. Bjornsson E, Kalaitzakis E, Olsson R. The impact of eosinophilia and hepatic necrosis on
prognosis in patients with drug-induced liver injury. Aliment Pharmacol Ther 2007;25:1411-1421.
104. Sasaki E, Matsuo K, Iida A, et al. A novel mouse model for phenytoin-induced liver injury:
involvement of immune-related factors and P450-mediated metabolism. Toxicol Sci 2013;136:250-263.
105. Pellock JM. Carbamazepine side effects in children and adults. Epilepsia 1987;28 Suppl 3:S64-
70.
106. Kalapos MP. Carbamazepine-provoked hepatotoxicity and possible aetiopathological role of
glutathione in the events. Retrospective review of old data and call for new investigation. Adverse Drug
React Toxicol Rev 2002;21:123-141.
107. Gokce S, Durmaz O, Celtik C, Aydogan A, Gulluoglu M, Sokucu S. Valproic acid-associated
vanishing bile duct syndrome. J Child Neurol 2010;25:909-911.
108. Bhayana H, Appasani S, Thapa BR, Das A, Singh K. Lamotrigine-induced vanishing bile duct
syndrome in a child. J Pediatr Gastroenterol Nutr 2012;55:e147-148.
109. Pellock JM. Felbamate in epilepsy therapy: evaluating the risks. Drug Saf 1999;21:225-239.
110. Pellock JM, Faught E, Leppik IE, Shinnar S, Zupanc ML. Felbamate: consensus of current
clinical experience. Epilepsy Res 2006;71:89-101.
111. Planjar-Prvan M, Bielen A, Sruk A, Marusic M, Bielen I. Acute oxcarbazepine-induced
hepatotoxicity in a patient susceptible to developing drug-induced liver injury. Coll Antropol
2013;37:281-284.
112. Chait Mermelstein A, Mermelstein J, Adam T, Brody BD, Dubin MJ. Oxcarbazepine-induced
liver injury after sensitization by valproic acid: a case report. Bipolar Dis 2016;18:307-309.
113. Bosdure E, Cano A, Roquelaure B, et al. [Oxcarbazepine and DRESS syndrome: a paediatric
cause of acute liver failure]. Arch Pediatr 2004;11:1073-1077.
114. Bjoro K, Gjerstad L, Bentdal O, Osnes S, Schrumpf E. Topiramate and fulminant liver failure.
Lancet 1998;352:1119.
115. Tsien MZ, Cordova J, Qadir A, Zhao L, Hart J, Azzam R. Topiramate-induced acute liver failure
in a pediatric patient: a case report and review of literature. J Pediatr Gastroenterol Nutr 2016;63:e37-38.
116. Li AM, Nelson EA, Hon EK, et al. Hepatic failure in a child with anti-epileptic hypersensitivity
syndrome. J Pediatr Child Health 2005;41:218-220.
117. Roberts EA, Spielberg SP, Goldbach M, Phillips MJ. Phenobarbital hepatotoxicity in an 8-month-
old infant. J Hepatol 1990;10:235-239.
118. Di Mizio G, Gambardella A, Labate A, Perna A, Ricci P, Quattrone A. Hepatonecrosis and
cholangitis related to long-term phenobarbital therapy: an autopsy report of two patients. Seizure
2007;16:653-656.
119. Santos NA, Medina WS, Martins NM, Rodrigues MA, Curti C, Santos AC. Involvement of
oxidative stress in the hepatotoxicity induced by aromatic antiepileptic drugs. Toxicol In Vitro
2008;22:1820-1824.

Page 26 of 37
120. Fayad M, Choueiri R, Mikati M. Potential hepatotoxicity of lamotrigine. Pediatr Neurol
2000;22:49-52.
121. Im SG, Yoo SH, Park YM, et al. Liver dysfunction induced by systemic hypersensitivity reaction
to lamotrigine: case report. Clin Mol Hepatol 2015;21:180-182.
122. Mecarelli O, Pulitano P, Mingoia M, et al. Acute hepatitis associated with lamotrigine and
managed with the molecular adsorbents recirculating system (Mars). Epilepsia 2005;46:1687-1689.
123. Broli M, Provini F, Naldi I, et al. Unexpected gamma glutamyltransferase rise increase during
levetiracetam monotherapy. Epileptic Dis 2010;12:81-82.
124. Syed AA, Adams CD. Acute liver failure following levetiracetam therapy for seizure prophylaxis
in traumatic brain injury. Case Rep Clin Med 2012;1:41-44.
125. Tan TC, de Boer BW, Mitchell A, et al. Levetiracetam as a possible cause of fulminant liver
failure. Neurology 2008;71:685-686.
126. Skopp G, Schmitt HP, Pedal I. [Fulminant liver failure in a patient on carbamazepine and
levetiracetam treatment associated with status epilepticus]. Arch Kriminol 2006;217:161-175.
127. Selvaraj V, Madabushi JS, Gunasekar P, Singh SP. Levetiracetam associated acute hepatic failure
requiring liver transplantation: case report. J Neurol 2016;263:814-815.
128. Sunwoo JS, Byun JI, Lee SK. A case of lacosamide-induced hepatotoxicity. Int J Clin Pharmacol
Ther 2015;53:471-473.
129. Gutierrez-Grobe Y, Bahena-Gonzalez JA, Herrera-Gomar M, Mendoza-Diaz P, Garcia-Lopez S,
Gonzalez-Chon O. Acute liver failure associated with levetiracetam and lacosamide combination
treatment for unspecified epileptic disorder. Case Rep Emerg Med 2013;2013:634174.
130. Ben-Menachem E, Biton V, Jatuzis D, Abou-Khalil B, Doty P, Rudd GD. Efficacy and safety of
oral lacosamide as adjunctive therapy in adults with partial-onset seizures. Epilepsia 2007;48:1308-1317.
131. Vuppalanchi R, Chalasani N, Saxena R. Restoration of bile ducts in drug-induced vanishing bile
duct syndrome due to zonisamide. Am J Surg Pathol 2006;30:1619-1623.
132. Coelho R, Rodrigues S, Gaspar R, Silva R, Lopes J, Macedo G. Zonisamide-induced Acute-on-
Chronic Liver Failure: First Report. J Clin Gastroenterol 2016;50:266.
133. Conilleau V, Dompmartin A, Verneuil L, Michel M, Leroy D. Hypersensitivity syndrome due to
2 anticonvulsant drugs. Contact Dermatitis 1999;41:141-144.
134. Coulter DL. Ethosuximide-induced liver dysfunction. Arch Neurol 1983;40:393-394.
135. Dossing M, Andreasen PB. Drug-induced liver disease in Denmark. An analysis of 572 cases of
hepatotoxicity reported to the Danish Board of Adverse Reactions to Drugs. Scand J Gastroenterol
1982;17:205-211.
136. Cunningham ML. Acute hepatic necrosis following treatment with amitriptyline and diazepam.
Br J Psychiatry 1965;111:1107-1109.
137. Stacher G. Intrahepatic cholostasis following combined diazepam-barbiturate therapy in patients
with tetanus. Wien Klin Wochenschr 1973;85:401-406.
138. Richardson CE, Williams DW, Kingham JG. Gabapentin induced cholestasis. BMJ
2002;325:635.
139. Bureau C, Poirson H, Peron JM, Vinel JP. [Gabapentine-induced acute hepatitis]. Gastroenterol
Clin Biol 2003;27:1169-1170.
140. Lasso-de-la-Vega MC, Zapater P, Such J, Perez-Mateo M, Horga JF. Gabapentin-associated
hepatotoxicity. Am J Gastroenterol 2001;96:3460-3462.
141. Ragucci MV, Cohen JM. Gabapentin-induced hypersensitivity syndrome. Clin Neuropharmacol
2001;24:103-105.
142. Sendra JM, Junyent TT, Pellicer MJ. Pregabalin-induced hepatotoxicity. Ann Pharmacother
2011;45:e32.
143. Dogan S, Ozberk S, Yurci A. Pregabalin-induced hepatotoxicity. Eur J Gastroenterol Hepatol
2011;23:628.
144. Crespo Perez L, Moreira Vicente V, Manzano Fernandez R, Garcia Aguilera XA. [Cholestasis
associated with pregabalin treatment]. Med Clin (Barc) 2008;130:157-158.

Page 27 of 37
145. Bamanikar A, Dhobale S, Lokwani S. Pregabalin hypersensitivity in a patient treated for
postherpetic neuralgia. Ind J Pharmacol 2013;45:522-523.
146. Locher C, Zafrani ES, Dhumeaux D, Mallat A. [Vigabatrin-induced cytolytic hepatitis].
Gastroenterol Clin Biol 2001;25:556-557.
147. Kellermann K, Soditt V, Rambeck B, Klinge O. Fatal hepatotoxicity in a child treated with
vigabatrin. Acta Neurol Scand 1996;93:380-381.
148. Massot A, Gimenez-Arnau A. Cutaneous adverse drug reaction type erythema multiforme major
induced by eslicarbazepine. J Pharmacol Pharmacother 2014;5:271-274.
149. Perrott J, Murphy NG, Zed PJ. L-carnitine for acute valproic acid overdose: a systematic review
of published cases. Ann Pharmacother 2010;44:1287-1293.
150. Russell S. Carnitine as an antidote for acute valproate toxicity in children. Curr Opin Pediatr
2007;19:206-210.
151. Chan YC, Tse ML, Lau FL. Two cases of valproic acid poisoning treated with L-carnitine.
Human Exp Toxicol 2007;26:967-969.
152. Lheureux PE, Hantson P. Carnitine in the treatment of valproic acid-induced toxicity. Clin
Toxicol (Phila) 2009;47:101-111.
153. Tsai MF, Chen CY. Valproate-induced hyperammonemic encephalopathy treated by
hemodialysis. Ren Fail 2008;30:822-824.
154. Lee WM, Hynan LS, Rossaro L, et al. Intravenous N-acetylcysteine improves transplant-free
survival in early stage non-acetaminophen acute liver failure. Gastroenterology 2009;137:856-864, 864
e851.
155. Sen S, Ratnaraj N, Davies NA, et al. Treatment of phenytoin toxicity by the molecular adsorbents
recirculating system (MARS). Epilepsia 2003;44:265-267.
156. Squires RH, Dhawan A, Alonso E, et al. Intravenous N-acetylcysteine in pediatric patients with
nonacetaminophen acute liver failure: a placebo-controlled clinical trial. Hepatology 2013;57:1542-1549.
157. Pischik E, Kauppinen R. An update of clinical management of acute intermittent porphyria. Appl
Clin Genet 2015;8:201-214.
158. Bylesjo I, Forsgren L, Lithner F, Boman K. Epidemiology and clinical characteristics of seizures
in patients with acute intermittent porphyria. Epilepsia 1996;37:230-235.
159. Solinas C, Vajda FJ. Epilepsy and porphyria: new perspectives. J Clin Neurosci 2004;11:356-361.
160. Bonkowsky HL, Sinclair PR, Emery S, Sinclair JF. Seizure management in acute hepatic
porphyria: risks of valproate and clonazepam. Neurology 1980;30:588-592.
161. Larson AW, Wasserstrom WR, Felsher BF, Chih JC. Posttraumatic epilepsy and acute
intermittent porphyria: effects of phenytoin, carbamazepine, and clonazepam. Neurology 1978;28:824-
828.
162. Doss M, Schafer HJ. Carbamazepine-induced acute porphyria syndrome. Lancet 1984;1:1026.
163. Rideout JM, Wright DJ, Lim CK, Rinsler MG, Peters TJ. Carbamazepine-induced non-hereditary
acute porphyria. Lancet 1983;2:464.
164. Yalouris AG, Lyberatos C, Chikrigi H, et al. [Effect of starvation and phenobarbital on the
activity of liver uroporphyrinogen synthetase]. Ann Biol Clin 1991;49:401-403.
165. Reynolds NC, Jr., Miska RM. Safety of anticonvulsants in hepatic porphyrias. Neurology
1981;31:480-484.
166. Granick S. The induction in vitro of the synthesis of delta-aminolevulinic acid synthetase in
chemical porphyria: a response to certain drugs, sex hormones, and foreign chemicals. J Biol Chem
1966;241:1359-1375.
167. Hahn M, Gildemeister OS, Krauss GL, et al. Effects of new anticonvulsant medications on
porphyrin synthesis in cultured liver cells: potential implications for patients with acute porphyria.
Neurology 1997;49:97-106.
168. Krijt J, Krijtova H, Sanitrak J. Effect of tiagabine and topiramate on porphyrin metabolism in an
in vivo model of porphyria. Pharmacol Toxicol 2001;89:15-22.
169. Lotte J, Haberlandt E, Neubauer B, Staudt M, Kluger GJ. Bromide in patients with SCN1A-
mutations manifesting as Dravet syndrome. Neuropediatr 2012;43:17-21.

Page 28 of 37
170. Bowers GN, Jr., Onoroski M. Hyperchloremia and the incidence of bromism in 1990. Clin Chem
1990;36:1399-1403.
171. Thadani H, Deacon A, Peters T. Diagnosis and management of porphyria. BMJ 2000;320:1647-
1651.
172. Magnussen CR, Doherty JM, Hess RA, Tschudy DP. Grand mal seizures and acute intermittent
porphyria. The problem of differential diagnosis and treatment. Neurology 1975;25:121-125.
173. Gaida-Hommernick B, Rieck K, Runge U. Oxcarbazepine in focal epilepsy and hepatic
porphyria: a case report. Epilepsia 2001;42:793-795.
174. Sykes RM. Acute intermittent porphyria, seizures, and antiepileptic drugs: a report on a 3-year-
old Nigerian boy. Seizure 2001;10:64-66.
175. Suzuki A, Aso K, Ariyoshi C, Ishimaru M. Acute intermittent porphyria and epilepsy: safety of
clonazepam. Epilepsia 1992;33:108-111.
176. Gorchein A. Drug treatment in acute porphyria. Br J Clin Pharmacol 1997;44:427-434.
177. Hedera P. Update on the clinical management of Wilson's disease. Appl Clin Genet 2017;10:9-
19.
178. Prashanth LK, Sinha S, Taly AB, Mahadevan A, Vasudev MK, Shankar SK. Spectrum of
epilepsy in Wilson's disease with electroencephalographic, MR imaging and pathological correlates. J
Neurol Sci 2010;291:44-51.
179. Dening TR, Berrios GE, Walshe JM. Wilson's disease and epilepsy. Brain 1988;111 ( Pt 5):1139-
1155.
180. Turk-Boru U, Kocer A, Alp R, Gumus M. Status epilepticus in a case with wilson's disease
during D-pencillamine treatment. Swiss Med Wkly 2003;133:446-447.

Page 29 of 37
Figure 1

Page 30 of 37
Table 1: Parenteral benzodiazepines used in acute treatment

Medication Metabolism Half-life in Heathy Subjects Onset of Action Duration of


Action
*Diazepam Hepatic Parent compound: 33 to 45 hours; 1 to 3 minutes 15-30 minutes
Active metabolite-
Desmethyldiazepam: ~87 hours

Lorazepam Hepatic Approximately 14 to 17.8 hours Within 10 minutes 6-8 hours

Midazolam Hepatic 1.8 to 6.4 hours (mean ~3 hours) 3 to 5 minutes < 2 hours

*Diazepam has high lipid solubility, rapidly reaching the brain. However, it redistributes to peripheral tissues in 15-20 minutes,
diminishing its anticonvulsant properties.

Page 31 of 37
Table 2: Pharmacological characteristics of antiepileptic drugs and recommendations for their use in patients with hepatic disease
Medication Metabolism Half-Life Protein Potential for Recommended Association Recommended Dose Recommended Dose
Binding Drug Monitoring in with Changes: Mild to Moderate Changes: Severe Hepatic
Interactions‡ Patients with Liver Hepatotoxicity║ Hepatic Disease Disease
Disease*
Brivaracetam Hepatic (20 ~ 9 hours ≤ 20% Minimal Clinical No known Manufacturer Manufacturer
%) association recommendation for patients recommendation for
≥ 16 years of age: Child patients ≥ 16 years of age:
Pugh A/B: Initial: 25 mg Child Pugh C: Initial: 25
twice daily, up to a mg twice daily, up to a
maximum of 75 mg twice maximum of 75 mg twice
daily daily
Carbamazepine Hepatic Children: 8-14 hours 75-90% High Drug Level Well established NSR NSR
Adults: 12-17 hours
Eslicarbazepine Hepatic 13-20 hours < 40% Minimal Clinical/Consider Low NSR NSR
Drug Level
Ethosuximide Hepatic Children: ~30 hours Insignificant Intermediate Clinical/ Consider Low NSR NSR
Adults: 50-60 hours Drug Level
Felbamate Hepatic 20-23 hours 22-25% High Drug Level Well established Contraindicated by Contraindicated by
manufacturer manufacturer
recommendation recommendation
Gabapentin None Children: ~4.7 hours < 3% Minimal Clinical Low NSR NSR
Adults: 5-7 hours
Lacosamide Hepatic ~13 hours < 15% Minimal Clinical/Consider Low Manufacturer NSR
(~60%) Drug Level recommendation for adults:
Maximum daily dose of 300
mg/day ¥
Lamotrigine Mainly hepatic Children: ~19 hours ~ 55% High Drug Level Low Manufacturer Manufacturer
Adults: 25-33 hours recommendation: recommendation: Severe
Mild: None without ascites: Reduce
Moderate without ascites: initial, escalation, and
Reduce initial, escalation, maintenance doses by
and maintenance doses by ~25%. Adjust based on
~25%. Adjust based on clinical response.
clinical response. Severe impairment with
Moderate impairment with ascites: Reduce initial,
ascites: Reduce initial, escalation, and
escalation, and maintenance maintenance doses by
doses by ~50%. Adjust based ~50%. Adjust based on
on clinical response. clinical response.
Levetiracetam Primarily Children:~5.3-6 < 10% Minimal Clinical Low None Decrease dose by half
enzymatic hours Adults:
hydrolysis in 6-8 hours
the blood

Page 32 of 37
Oxcarbazepine Hepatic Active metabolite: Parent drug: Intermediate Clinical/Consider Low None NSR
Children: 4.8 to 9.3 67% Drug Level
hours Active
Adults: ~9 hours metabolite:
40%
Perampanel Hepatic ~105 hours 95-96% High Drug Level No known Manufacturer Manufacturer
association recommendation: recommendation: Child-
Child-Pugh class A: Pugh class C: NSR
Maximum daily dose: 6
mg/day Child-
Pugh class B: Maximum
daily dose: 4 mg/day
Phenobarbital Hepatic Children: ~110 hours 50-60% High Drug Level Low NSR NSR
(60-180)
Adults: ~79 hours
(53-118)
Phenytoin Hepatic 7-42 hours ≥85% High Drug Level Well established NSR NSR
Half-life increases
with increasing
phenytoin
concentrations
Pregabalin Negligible ~6.3 hours 0% Minimal Clinical Low NSR NSR
metabolism
Tiagabine Hepatic Children: ~5.7 hours 96% High Clinical/Consider No known NSR NSR
(2-10) Drug Level association
Adults: 7-9 hours
Topiramate Hepatic (20 Children: 7.7-12.8 15-41% † Minimal Clinical Low NSR Decrease by 30%
%) hours Adults:
19-23 hours
Valproic Acid Hepatic Children: 7-13 hours 80% - 90% High Drug Level Well established Not recommended by Contraindicated by
Adults: 9-19 hours manufacturer manufacturer
Vigabatrin No significant Children: 5.7-9.5 0% Minimal Clinical Low NSR NSR
metabolism hours
Adults:~10.5 hours
Zonisamide Hepatic (70%) ~69 hours 40% Minimal/ Clinical/Consider Low NSR NSR
Intermediate Drug Level
* Drug level is recommended for AEDs with high chance of drug-drug interactions or toxicity. Especially patients with polytherapy
† Protein binding capacity inversely related to plasma levels
‡ Drugs with high protein binding capacity and/or potential to inhibit or induce hepatic enzymes have high chances for drug-drug interaction
║Hepatotoxicity is a potential but rare side effect of AEDs
¥ Concentration of lacosamide can increase by 50-60% in patients with moderate liver dysfunction.
NSR=No specific recommendation

Page 33 of 37
Table 3: Child-Pugh Class Scores
Data derived from Trey, Burns, and Saunders, 1966, with reproduction permission from Wiley Global Permissions.

Measurement 1 Point 2 Points 3 Points


Bilirubin (total) mg/dL <2 2-3 >3
Serum albumin g/L >35 28-35 <28
INR <1.7 1.71-2.20 >2.20
Ascites None Mild Severe
Hepatic None Grade I-II (or Grade III-IV (or
encephalopathy suppressed with refractory)
medication)

Interpretation

Points Class One year survival Two year survival


5-6 A 100% 85%
7-9 B 81% 57%
10-15 C 45% 35%

Page 34 of 37
Table 4: Calculated risk of hepatotoxicity for specific antiepileptic drugs

Medication Calculated risk Identified or potential risk factors

Carbamazepine Rare Children with polytherapy

Felbamate 1 per 26,000 to 1 per 34,000 Female gender


Polytherapy

Phenytoin 1 per 10,000 to 1 per 50,000 Black ethnicity


Children less susceptible

Valproic Acid Monotherapy Polymerase gamma 1 (POLG1) mutations


Adults: 1 per 45,000 Younger Age
Children < 2 years: 1 per 7,000 Polytherapy
Developmental delay
Polytherapy Metabolic disorders
Adults: 1 per 12,000 Febrile illness
Children < 2 years: 1 per 500-600 Status epilepticus

Page 35 of 37
Table 5: Treatment modalities used in cases of hepatotoxicity associated with specific AEDs

Medication Treatment Notes


Valproic acid L-carnitine Doses of 100 mg/kg followed intravenous doses of
50 mg/kg every 8 hours, to a maximum of 3 grams
per dose.149-152
Hemodialysis, Used for progressive neurological deterioration or
Sodium benzoate ammonia levels greater than 680 μg/dL (400
phenyl acetate μmol/L).153
Carbamazepine N- acetylcysteine * Doses used are 150 mg/kg/hour over one hour,
followed by 12.5 mg/kg/hour for 4 hours, then
continuous infusions of 6.25 mg/kg/hour for the
remaining 67 hours.154
Steroids Hypersensitivity reactions
Phenytoin N- acetylcysteine Doses used are 150 mg/kg/hour over one hour,
followed by 12.5 mg/kg/hour for 4 hours, then
continuous infusions of 6.25 mg/kg/hour for the
remaining 67 hours.154
Molecular adsorbents Used to remove molecules that are highly protein
recirculating system bound.122, 155
device (MARS)
Steroids Hypersensitivity reactions

Lamotrigine Molecular adsorbents Used to remove molecules that are highly protein
recirculating system bound.122, 147
device (MARS)
Steroids Hypersensitivity reactions

* N- acetylcysteine and antidote for acetaminophen DILI has recently showed benefit for adult
patients with non-acetaminophen DILI, including cases of carbamazepine and phenytoin
hepatotoxicity. On the contrary, N- acetylcysteine was of no benefit in young children with non-
acetaminophen acute liver failure.

Page 36 of 37
Table 6: Potassium Bromide* dosing and potential side effects

Initial dose Maintenance dose Half-life Common side Rare side effects
effects
30 30-80 mg/kg/day Children: 6-8 Drowsiness Bromism
mg/kg/day Increase by 10 days Gastrointestinal (Psychiatric
mg/kg/day every 2 Adults: 8-14 Acneiform symptoms,
weeks days eruption nausea, vomiting,
Concentration rash)172 †
impairment
Behavioral171
*Other formulations (ammonium bromide/potassium bromide/sodium bromide) exist and may
have different dosing regimens; †An incidence of 1 in 1,000,000 has been estimated

Page 37 of 37

You might also like