Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/339259258

Therapeutic drug monitoring of antiepileptic drugs: current status and future


prospects

Article · February 2020


DOI: 10.1080/17425255.2020.1724956

CITATIONS READS

90 3,787

3 authors:

Cecilie Johannessen Landmark Svein Johannessen


Oslo Metropolitan University and The National Center for Epilepsy and Dept of Ph… The National Center for Epilepsy, Oslo University Hospital, Oslo, Norway
103 PUBLICATIONS 3,480 CITATIONS 177 PUBLICATIONS 8,728 CITATIONS

SEE PROFILE SEE PROFILE

Philip Patsalos
University College London
435 PUBLICATIONS 14,156 CITATIONS

SEE PROFILE

All content following this page was uploaded by Philip Patsalos on 17 June 2020.

The user has requested enhancement of the downloaded file.


EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY
https://doi.org/10.1080/17425255.2020.1724956

REVIEW

Therapeutic drug monitoring of antiepileptic drugs: current status and future


prospects
Cecilie Johannessen Landmarka,b,c, Svein I. Johannessenb,c and Philip N. Patsalosd
a
Program for Pharmacy, Department of Life Sciences and Health, Faculty of Health Sciences, Metropolitan University, Oslo, Norway; bThe National
Center for Epilepsy, Sandvika, Oslo University Hospital, Oslo, Norway; cSection for Clinical Pharmacology, Department of Pharmacology, Oslo
University Hospital, Oslo, Norway; dDepartment of Clinical & Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK

ABSTRACT ARTICLE HISTORY


Introduction: Antiepileptic drugs (AEDs) are the cornerstone of treatment of patients with epilepsy, and Received 13 November 2019
there are presently 27 licensed AEDs making AEDs among the most common medications for which Accepted 29 January 2020
therapeutic drug monitoring (TDM) is performed. The aim of this review is to provide an overview of the KEYWORDS
current evidence of the use and implementation of AED TDM in patients with epilepsy and other non- Antiepileptic drugs; epilepsy;
epilepsy conditions. drug interactions;
Areas covered: The pharmacokinetic variability of AEDs is extensive, resulting in pronounced variability pharmacokinetic variability;
in serum concentrations between patients. TDM may thus be useful to individualize the treatment of therapeutic drug monitoring
patients with epilepsy and also in non-epilepsy conditions. Indications for TDM include settings where
pharmacokinetic variability is anticipated (e.g. in children, the elderly, during pregnancy, and patients
prescribed polytherapy resulting in drug interactions) and drug adherence. TDM contributes to provide
a quality assurance of the treatment. Patient management is, therefore, best guided by the determina-
tion of individual therapeutic concentrations.
Expert opinion: Because of pharmacokinetic variability is prevalent among AEDs, TDM allows
a bespoke approach to epilepsy care allowing dose adjustments based on measured drug concentra-
tions so as to optimize clinical outcome. Future advances include the use of additional markers of
toxicity and genetic variability so as to further aid individualization and optimize AED treatment.

1. Introduction in a prolonged effect in the brain, even though the serum


concentration of vigabatrin is declining or even zero.
Therapeutic drug monitoring (TDM) is defined as the mea-
AEDs serve as the cornerstone of treatment of patients with
surement and clinical use of serum/plasma (or saliva) drug
epilepsy. In recent years, several new AEDs have been intro-
concentrations to adjust each patient’s individual dosage
duced to the market, and 27 AEDs are available internationally
and thus schedule to each patient’s individual therapeutic
[1]. For all AEDs, there is pronounced pharmacological varia-
requirement [1–8]. Although few randomized studies have
bility, where at present pharmacokinetic variability is most
demonstrated a positive impact of TDM on the clinical out-
easily measurable and might be controlled for [3,4].
come in epilepsy, evidence from non-randomized studies
Environmental, physiological, and genetic factors also contri-
and everyday clinical experience indicates that the use of
bute to extensive variability in the obtained serum concentra-
TDM for older as well as newer antiepileptic drugs (AEDs)
tions of any given AED. Also, aspects of adherence, if the drug
may best be used to guide patient management, provided
is taken as prescribed, come into play in the total evaluation of
that serum concentrations are measured with a clear indica-
the AED treatment.
tion and with a clinical interpretation [6]. AED TDM has
The onset of epilepsy is predominantly in early life, it is
been used to manage pharmacological variability in and
a chronic condition that often lasts for years or the whole life-
between patients for the last 50 years, and many drugs
time, and many patients require long-term therapy. The principal
have become available during that time, allowing continu-
outcome in epilepsy is the absence of seizures. Two-thirds of
ous development of this field of research and evaluating its
patients achieve seizure control with monotherapy AEDs, and
impact on clinical practice. TDM is relevant for all drugs
20–30% require polytherapy [2,9,10]. Furthermore, even though
where the serum concentration is supposed to reflect the
there are now numerous available AEDs, the number of patients
concentration and pharmacological action at target in the
that are not responding to a first AED is 50%, and one-third is
brain. The only exception to this is vigabatrin, which is an
still regarded as being treatment resistant, many of which have
irreversible inhibitor of the enzyme responsible for the
several seizure types [10,11]. Indeed, the choice of the appro-
degradation of GABA, GABA transaminase. This can result
priate AED for different seizure types is of paramount

CONTACT Cecilie Johannessen Landmark cecilie.landmark@hioa.no Program for Pharmacy, Oslo Metropolitan University and the National Center for
Epilepsy, Pilestredet 50, N-0167 Oslo/G. F. Henriksens Vei 23, Sandvika, Oslo N-1300, Norway and Deparment of Pharmacology, Oslo University Hospital, Pilestredet
50, N-0167 Oslo/G. F. Henriksens Vei 23, Sandvika, Oslo N-1300, Norway
© 2020 Informa UK Limited, trading as Taylor & Francis Group
2 C. JOHANNESSEN LANDMARK ET AL.

variability in general and in special patient groups, drug inter-


Article highlights actions, and pharmacogenetic variability.
● The pharmacokinetic variability of all AEDs is extensive, resulting in
pronounced variability in serum concentrations between patients,
and may be challenging in special patient groups such as children,
3.1. Pharmacokinetic variability in special patient
pregnant women, and the elderly populations
● Therapeutic drug monitoring (TDM) contributes to quality assurance
of the treatment, taking pharmacokinetic variability, drug interac- During the transition from childhood, adolescence, adulthood,
tions, adherence, and other treatment challenges into account and further to old age, significant changes in physiology and
● Future advances for TDM of AEDs include use of additional markers of
toxicity and genetic variability to individualize and optimize
pathology occur, such as hepatic and renal function, uremia,
treatment and other diseases contributing to changes in the pharmaco-
kinetic characteristics of AEDs [1–6] (see Tables 1 and 2).
This box summarizes key points contained in the article.

3.1.1. Infants and children


In children, physiological alterations cause rapid changes in
pharmacokinetics during early childhood. For most AEDs that
importance, as some AEDs are specifically effective in certain
have been studied in the youngest children, the clearance is
seizure types, for example, ethosuximide in absence seizures and
high, and thus, the elimination half-life is low, especially from
stiripentol in Dravet syndrome [12,13]. Thus, in patients with
6 months to about 6 years of age. The volume of distribution
refractory epilepsy with a challenging treatment and seizure
also changes [3,14–16]. The clinical impact is that infants and
occurrence, TDM may be a valuable tool to optimize and indivi-
young children often need a higher dosage per kilogram body
dualize AED treatment and to monitor treatment over time.
weight than older children and adolescents. This extensive
The aim of this review is to provide an updated overview of
pharmacokinetic variability during development makes it dif-
the current evidence of the use and implementation of TDM in
ficult to predict optimal therapeutic doses, and TDM may be
epilepsy and other indications. Reasons for TDM will be high-
particularly helpful in these patient groups. In older children
lighted, such as pharmacokinetic variability, drug interactions,
and adolescents, the pharmacokinetics are similar to that in
adherence, and other treatment challenges, where TDM con-
adult populations.
tributes to provide a quality assurance of the treatment.
Furthermore, key principles relating to the appropriate inter-
3.1.2. Women during pregnancy
pretation of the reference range and individual reference con-
Physiological processes change in pregnancy, and these
centrations, matrix selection, and analytical aspects will be
changes lead to decreased absorption, altered distribution,
discussed. Overall, we aim at presenting evidence-based
decreased protein binding of highly bound drugs, increased
knowledge allowing appropriate and optimal implementation
metabolism due to increased enzyme capacity of cytochrome
of TDM in clinical settings.
P450 (CYP) and uridine glucuronyl transferase (UGT) isoen-
zymes, and increased excretion [2,17,18]. The result is an
2. Search strategy and selection criteria increase in serum concentration/dose-ratio from up to 300%
for lamotrigine to 30–50% for other newer AEDs including
This study is a review based on published articles and searches levetiracetam, oxcarbazepine, topiramate, and zonisamide,
in PubMed and Google Scholar, up to January 2020, with during pregnancy and with rapid changes back to baseline
a focus on recent advances from the last 10 years. Peer- post-partum [17–26]. Gabapentin, now most relevant for
reviewed articles in internationally recognized journals or women with neuropathic pain, also decreases in serum con-
scientific books written in English are included, and primary centrations during pregnancy [27]. The use of valproate is now
sources were preferred. The included search terms included contraindicated during pregnancy unless other alternatives
one or more of the following, alone or in combination: anti- have failed due to increased risk of teratogenic and long-
epileptic drugs, and all the individual AEDs: brivaracetam, term cognitive development and autism-spectrum disorders
carbamazepine, carbamazepine-epoxide, clobazam, clonaze- in the offspring [28–30]. The pharmacokinetic variability of
pam, diazepam, eslicarbazepine acetate, ethosuximide, felba- valproate in women is extensive, and for a safe management
mate, fosphenytoin, gabapentin, lacosamide, lamotrigine, of highly protein-bound drugs like valproate, it might best be
levetiracetam, oxcarbazepine, perampanel, phenobarbital, guided by measurement of free, unbound, concentrations
phenytoin, pregabalin, primidone, rufinamide, stiripentol, [31,32].
sulthiame, tiagabine, topiramate, valproic acid, vigabatrin,
and zonisamide. Other terms included cytochrome P-450, 3.1.3. The elderly and patients with hepatic and renal
dried blood spots, drug interactions, epilepsy, pharmacology, impairment
pharmacokinetics, pharmacokinetic variability, pharmacoge- The clearance of all AEDs is reduced by 30–50% in the elderly, and
netics, precision medicine, serum or plasma, and TDM. in some cases, even greater changes with up to 90% have been
observed [15]. Pronounced physiologic changes affect the phar-
macokinetic processes in the body and thus characteristics of
3. Pharmacological variability of AEDs
AEDs, such as decreased absorption, altered distribution, and
Factors contributing to pharmacological variability within and decreased metabolic capacity, blood flow to eliminating organs,
between patients are described, such as pharmacokinetic and thus hepatic and renal clearance [3,6]. Polypharmacy is
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 3

Table 1. Indications for TDM and clinical consequences. common in the elderly, with up to nine concomitantly prescribed
Clinical consequence and guidance in CNS-active drugs being reported [9]. The elderly may show
Indications for TDM further therapy increased pharmacodynamic sensitivity to AEDs and other drugs,
When a drug is initiated, or dosage Determine the serum concentration as and therapeutic and toxic effects may occur at even low concen-
or formulation is changed a guide to further follow up and
confirm that the change in serum trations. Interpretation of the TDM results should, therefore, be
concentration is reflected by the evaluated more cautiously.
change in dosage. This is of As most AEDs are metabolized by the liver, hepatic disease
particular importance for AEDs with
saturation kinetics (phenytoin, may alter clearance [3,6]. Renal function is especially relevant
stiripentol) and non-linear kinetics for AEDs eliminated partly or mainly by renal excretion, such
(carbamazepine, valproic acid, as pregabalin, gabapentin, levetiracetam, lacosamide, and vig-
gabapentin). Following a change of
formulation, Cmax and Tmax may abatrin [3,6]. Many AEDs are efficiently removed during dialy-
differ. sis but may be challenging to handle, and TDM may help
When an optimal therapeutic Establish an ‘individual therapeutic guide replacement dosages to maintain seizure control. In
outcome is reached concentration’ of the AED as
a guide for future changes in the patients with hepatic and renal diseases, TDM is recom-
therapy mended, and for highly bound AEDs, the free, unbound con-
When adverse effects occur Confirm whether the serum centrations may better guide treatment, as the change in
concentration is high and might be
a probable cause of drug toxicity. protein binding and clearance is unpredictable and few stu-
This is of special importance for dies have been performed.
drugs with a rapid absorption and
high Cmax values such as
oxcarbazepine or benzodiazepines. 3.2. Drug interactions
If nonadherence is suspected Serial serum concentrations when an
individual reference concentration In epilepsy, monotherapy is preferred, but since one-third of
has been established may reveal
variable adherence that should be patients are suffering from refractory epilepsy, polypharmacy
discussed with the patient with several AEDs is often necessary. A challenge in the man-
At therapeutic failure To establish whether a lack of efficacy agement of polypharmacy is pharmacokinetic interactions, as
may be due to a suboptimal dose/
serum concentration or if the dose/ there are numerous interactions between AEDs, where potent
serum concentration is too high enzyme inducers and inhibitors are among the most com-
and may produce a paradoxical monly used AEDs (for review, see [1,4,33–37]).
worsening of seizures, or if poor
adherence is suspected Pharmacokinetic interactions may be controlled for by the
Individual variations between dose Pharmacokinetic or pharmacogenetic use of TDM since changes in serum concentrations are
and serum concentrations variability, ethnicity, age, a consequence of changes in metabolism [2–4,6]. When an
pharmacoresistance, concurrent
illness, and organ function (liver, interacting drug is withdrawn, the interaction will be reversed,
kidneys) are among factors that and thus, it is just as important to adjust for any dosage
contribute to inter-individual changes made accordingly, and this is best guided by TDM.
variability and which applies to all
AEDs. Pharmacogenetic variability Enzyme induction leads to decreased serum concentrations
may, for instance, be identified by and an increased risk of seizure breakthrough. Enzyme induc-
unexpectedly high ratios between tion is completed within 2–3 weeks, and therefore, serum
clobazam and the active metabolite
desmethylclobazam (due to concentration measurements should be performed at this
CYP2C19 polymorphism). point. Enzyme inhibition, on the other hand, may give rise to
Pharmacokinetic variability in special Children, pregnant women, and the increased serum concentrations and excessive adverse effects
patient populations elderly have different dose
requirements due to alteration in or toxicity and implies that a new steady-state serum concen-
physiology and consequently AED tration is achieved, depending on the half-life of the affected
pharmacokinetics drug and takes about five half-lives after the inhibiting drug
Pharmacokinetic interactions Pharmacokinetic interactions are
common in epilepsy and result in has been initiated. The clinical consequence of an interaction
increases or decreases in AED may be difficult to predict in the individual patient due to
serum concentrations with other patient-specific features. Thus, TDM may be used to
potential occurrence of adverse
effects or seizure breakthrough, determine the magnitude of the interactions and make proper
respectively. AED interactions dosage adjustments, with an individual therapeutic concentra-
involving non-AEDs are also tion of the patient as a baseline.
prominent.
Emergency situations: overdose, To guide when the AED dose should Knowledge of the mechanism of interactions helps in decision-
status epilepticus, acute be reinstated to maintain seizure making, as no database is complete or updated at all times. For
pathological states (e.g. stroke, control. Determine possible instance, valproate, stiripentol, and felbamate are potent enzyme
surgery, head trauma) nonadherence and guide a suitable
dosage regimen to control seizures. inhibitors of certain CYPs and UGT enzymes and affect the serum
Acute changes in pharmacokinetics, concentrations of other AEDs [1–6,35–37]. Typical enzyme indu-
for example in protein binding, cers include carbamazepine, phenytoin, and phenobarbital
may occur.
(including primidone), acting on several hepatic enzymes, but
4 C. JOHANNESSEN LANDMARK ET AL.

Table 2. Useful pharmacokinetic characteristics of AEDs so as to aid the implementation and interpretation of TDM data.
Time to
peak Time to
conc. steady Half-life, Half-life, Half-life, Protein Route of elimination Reference Reference
Drug (h) state (days) adults (h) children (h) elderly (h) binding (%) (mainly hepatic) range (mg/L) range (μmol/L)
First-generation antiepileptic drugs
Carbamazepine 4–8a 2–4 8–20b 10–13b 30–50b 75 CYP1A2, 2C8, 3A4 4–12 17–51
Clobazam Desmethyl 1–3 2–7 10–30 ~16 30–48 90 CYP3A4 0.03–0.3 0.1–1.0
clobazam (metabolite) 7–10 36–46 2C19 0.02–0.07 1.0–10.5
Clonazepam 1–4 2–10 17–56 22–33 ____ 90 CYP2B, 2E1, 3A4 0.02–0.07 0.06–0.22
Ethosuximide 1–4 8–12 40–60 30–40 ____ 22 CYP2B, 2E1, 3A4 40–100 283–708
Phenobarbital 2–4 15–29 70–140 63–69 ____ 48 CYP2E1, 2C19 10–40 43–172
Phenytoin 1–12 6–21 30–100b 30–100b ____ 92 CYP2C9, 2C19 10–20 40–79
Primidone 2–6 2–4 7–22 5–11 ____ 33 CYP2E1, 2C9/19? 5–10 23–46
d
Valproic acid 3–7 2–4 12–16b 8–13b ____ 74–93d CYP2A6, 2C9/19, 2B6, 50–100 346–693
UGT1A3, 2B7
Second-generation antiepileptic drugs
Felbamate 2–6 3–5 16–22 ____ ____ 48 CYP3A4, 2E1 30–60 126–252
Gabapentin 2–3 1–2 5–9b ____ ____ 0 No, renal 2–20 12–117
Lamotrigine 1–3 3–7 15–35 ____ ____ 66 UGT1A4, 2B7 3–15 10–59
Levetiracetam 1–2 1–2 6–8 5–6 10–11 3 Type B esterase 12–46 70–270
c
Oxcarbazepine 4–6 2–3 8–15 ____ ____ 40 Arylketone reductase, 3–35 12–139
UGTs
Pregabalin 1–2 1–2 5–7 ____ ____ 0 No, renal 2–8 13–50
Tiagabine 0.5–2 1–2 5–9 ____ ____ 98 CYP3A4 0.02–0.2 0.05–0.53c
Topiramate 2–4 4–5 20–30 13–20 ____ 20 CYP3A4 5–20 15–59
Vigabatrin 1–2 1–2 5–8 ____ ____ 17 No, renal 2–36 6–279
Zonisamide 2–5 10–15 50–70 ____ ____ 44 CYP3A4 10–40 47–188
Third-generation antiepileptic drugs
Brivaracetam 0.5–2 2 7–8 ____ ____ 35 Hydrolysis, CYP2C19 0.2–2.0 1–10.0
Eslicarbazepinec 2–3 4–5 20–24 ____ ____ 44 Esterases, UGTs, ? 3–35 12–139
Lacosamide 1–2 2–3 12–16 ____ ____ 14 CYP2C19 10–20 40–80
Perampanel 0.25–2 14 52–129 ____ ____ 98 CYP3A4 0.05–0.4 0.14–1.14
Rufinamide 3–6 1–2 6–10 ____ ____ 28 Hydrolysis, non-CYP 30–40 126–168
dependent
Stiripentol 1–2 7 4–13b ____ ____ 96 CYP1A2, 2C19, 3A4 4–22 17–94
a
Conventional tablets; Nonlinear pharmacokinetics t; cValues refer to pharmacologically active metabolite licarbazepine (10-hydroxycarbazepine); dEnteric-coated
b

tablets. Half-lives are based on monotherapy. N.E. = not established. Based on [2,3,6,11,12].

the latter is weaker as an inducer on CYP3A4 [38]. Carbamazepine 3.3. Pharmacogenetic variability in metabolizing
undergoes autoinduction so that its clearance can increase three- enzymes
fold within several weeks of starting therapy; also, the formation of
Genetic polymorphisms in drug-metabolizing enzymes, such
the active epoxide metabolite varies over time [39,40].
as CYP2C9, CYP2C19, and CYP4CA, also contribute to pharma-
Consequently, it is important to undertake TDM after autoinduc-
cological variability of AEDs. CYP2D6, a common metabolizing
tion has completed.
isoenzyme, is not involved in AED metabolism. These muta-
Lamotrigine is for instance affected by both enzyme inducers
tions may be identified by genotyping [3,51]. Simple kits for
and inhibitors [41,42]. In some cases, pharmacokinetic interactions
CYP genotyping are available as specific CYP kits or as part of
and pharmacogenetic variability in metabolizing enzymes may be
broader biochemical screening kits. This could be useful as
revealed by the measurement of the drug and metabolite, such as
a-priori testing for certain drugs where mutations such as poor
for clobazam and the active metabolite desmethyl-clobazam
metabolizing phenotypes give rise to excessive adverse effects
[43,44]. Drugs that are only renally excreted are not likely to be
at therapeutic doses. Perhaps these kits could be used where
involved in pharmacokinetic interactions, such as gabapentin,
well-equipped TDM laboratories are not established. However,
pregabalin, and vigabatrin.
limitations might still be costs, precision, and evaluation of the
In addition to polypharmacy with AEDs, many patients
results. To assess the pharmacokinetic phenotype in patients,
suffer from comorbid psychiatric affects where additional
probe drug phenotype testing has been investigated for spe-
pharmacological treatment is needed such as antidepressants
cific CYP enzymes where pharmacogenetic variability plays
and antipsychotics and where interactions are likely to occur
a role, such as caffeine for CYP1A2 or omeprazole for
[9,45,46]. Other important disorders require the use of drug
CYP2C19. The value in clinical practice is, however, limited to
classes often involved in interactions, such as analgesics, anti-
the fact that few drugs are metabolized by a single CYP
microbials, antineoplastic agents, HIV drugs, immunosuppres-
isoenzyme only, and conventional TDM seems to be more
sants, cardioactive drugs including warfarin or newer oral
applicable [52].
anticoagulants, and steroids including oral contraceptives
However, in many cases, it is easier to monitor the serum
[36,44–50].
concentration and adjust the dose needed in the particular
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 5

patient. Indeed, it has recently been pointed out that, in micro-sampling tool has also recently been described which
clinical practice, genetic testing is only recommended in aids ease of use [64,65].
a very limited number of cases [53]. Then, additional phar-
macogenetic testing may elucidate the reason for unex-
4.2. What to measure? Total/unbound concentrations
pected relationships in serum concentrations and dose (C/
and pharmacologically active metabolites
D) of certain AEDs where CYP2C9 and 2C19 and, to some
extent, UGTs have polymorphisms, resulting in altered meta- For the majority of patients, the total plasma or serum AED
bolic capacity. Susceptible AEDs where pharmacogenetic concentration is measured [4,6]. However, for highly protein-
variability is a concern include clobazam, phenytoin, and, to bound AEDs (>90% bound, e.g. phenytoin and valproic acid
a lesser extent, lamotrigine [43,44,54]. For valproate, poly- and possibly perampanel, stiripentol, and clobazam), measure-
morphisms in these enzymes did not correlate to significant ment of free, unbound concentrations may be indicated, as
changes in C/D-ratios and does therefore not seem to be of albumin and thus protein binding are decreased, e.g. in the
clinical relevance [55]. elderly, those with decreased organ function or during preg-
Pharmacoresistance could be explained by genetic variabil- nancy. The consequence of increased unbound drug concen-
ity in the expression of drug transporters such as tration is adverse effects even at moderate total
P-glycoprotein among other hypotheses but is still under concentrations [7].
debate and needs further research [51,56]. An interesting Carbamazepine has a pharmacologically active metabolite
approach was recently demonstrated by using (carbamazepine-10,11-epoxide) that is measured in many
a bioinformatic in silico model for predicting dysfunctional laboratories because it is equipotent to carbamazepine and is
genes such as solute carrier genes and their contribution to commonly associated with adverse effects. The concentration
pharmacoresistance in epilepsy [57]. of the epoxide metabolite is highly variable and is dependent
on age, autoinduction of carbamazepine, and co-medication.
For example, valproate, phenytoin, phenobarbital, and brivara-
4. Principles for TDM
cetam increase the concentration of the epoxide and can cause
There are several reasons why it might be difficult to identify adverse effects [66–70]. Increased concentrations of the epox-
the optimal dose(s) in the individual patient on clinical ide could result in adverse effects. Based on these findings, the
grounds alone: (1) clinical effects of AEDs correlate better to concentration of the epoxide in the blood is referred to be
the serum concentration than to the dose due to extensive 5–15% of the parent drug [66,67].
pharmacokinetic variability both between and within patients; Clobazam also has an active metabolite, desmethyl-
(2) the nature of seizures is variable in intensity and intervals; clobazam, and its concentration in serum can be 10-fold
(3) variable adherence contributes to variable drug exposure; higher than the parent drug [43,44]. Experience has shown
(4) it might be difficult to recognize signs of toxicity on clinical that measurement of desmethyl-clobazam can be very useful
grounds; and (5) there are no laboratory markers for clinical as a marker of adverse effects in many patients [6,43,44,71].
efficacy or toxicity of AEDs [2-6].
4.3. Quality assurance and methodological aspects
4.1. Matrix for measurement: serum/plasma, saliva, and
For optimal quality assurance, the laboratories offering a TDM
dried blood spots
service should be part of an independent quality assurance
For AED TDM, serum or plasma is most commonly used, and program and use evaluated analytical methodologies. Typical
reference ranges are based on measurements of the total methodologies include HPLC/UPLC combined with UV- or MS/
concentration of the drug [2,6,7]. Saliva may also be employed MS detection and immunological methods. Increasingly, LC-
since concentrations in saliva reflect the pharmacologically MS is becoming the methodology of choice. Of course, the use
active serum unbound drug concentrations for several AEDs of such advanced methodologies requires highly qualified
[8]. Saliva sampling can be undertaken by patients or their staff with the ability not only to analyze samples but also to
caregivers and may be useful in children and other vulnerable undertake equipment maintenance.
patients. Sampling is simple and noninvasive, and moreover, To achieve comparable results from one measurement to
multiple samples may be collected. The sampling conditions another and between patients, the blood sampling time for
need to be standardized since the saliva concentrations of individual patients should be standardized. Trough concentra-
AEDs can be affected by contamination of mucus and the tions are preferred, and it should be noted that the various
presence of residual drug and food in the mouth [8]. If the reference ranges comprise trough concentrations [3–6,71]. It is
conditions are well controlled, saliva concentrations may be recommended that blood samples are drawn at steady state,
used to individualize AED treatment. This may be useful when and typically, this entails about five half-lives after initiating
protein binding is altered for highly protein-bound AEDs [2,6– treatment or following a dose adjustment. For all AEDs, the
8]. Dried blood spot analysis is an alternative technique and is ideal sampling time is in the morning, before intake of the
in increasing use consequent to the recent improvement in morning dose, representing the trough concentration. For
technology and analytical sensitivity [58,59]. It was, however, outpatients, the morning dose can be postponed for
highlighted in a recent review that more attention should be a couple of hours to obtain this. In the case of toxic symptoms,
drawn at the clinical validation of such methods [60]. Home- another sample may be drawn at the time when adverse
sampling in vulnerable patients is also possible [61–63]. A new effects occur to determine whether the symptoms are indeed
6 C. JOHANNESSEN LANDMARK ET AL.

drug related. Recently, postictal TDM has been shown to be a 7% reduction in costs for those with >50% seizure free-
useful after unexpected breakthrough seizures so as to assess dom and a 22% reduction in costs following a 3-month
adherence [72,73]. seizure-free period. The incremental cost-effectiveness
ratio was sensitive to the cost of management. The
authors call for longer follow-up periods in future studies
4.4. Cost-effectiveness [78]. In a recently published study, the economical burden
for caregivers was highlighted based on a questionnaire to
Few studies have been performed to assess the cost-
500 caregivers for persons with epilepsy in the US. It was
effectiveness of the use of AED TDM, and indeed, an
demonstrated that the burden was nearly 48 billion USD
overview of the cost-effectiveness of TDM for various
per year and greatest for those who care for children with
other therapeutic classes reports the same lack of studies
frequent seizures. Thus, the impact on caregivers also
[74]. Eadie reviewed 25 years of practice following the
should be considered when estimating the values of inter-
introduction of AED TDM and found a significantly higher
ventions to control epilepsy [79].
rate of seizure freedom in those who had access to TDM
during the first 6 months after their first seizure [75].
Furthermore, he discussed the possible cost-effectiveness 5. Implementation of TDM – current status
regarding a decreased need for drug dosage in some
patients and the lack of extra costs related to the devel- TDM provides bespoke AED treatment of patients with epilepsy.
opment of adverse effects [76]. Such studies are still lack- Table 1 highlights the indications for AED TDM, while Table 2 lists
ing to support these assumptions in terms of reduction in the various useful pharmacokinetic characteristics of AEDs so as
costs, hospitalization, or other outcome measures. Two to aid the implementation and interpretation of TDM data.
studies have been performed, where groups could be Figure 1 is a schematic of variables that need to be considered
divided to access to TDM or not, in India and Iraq when TDM is undertaken. The ultimate goal is to determine the
[77,78]. In the study from India, 25 patients who had patient’s individual AED therapeutic concentration and range,
been monitored with TDM and 25 without, and and this approach is useful for all AEDs.
a retrospective pharmacoeconomic analysis was performed
after 1 year. The outcome measures demonstrated signifi-
5.1. How to use the terminology of reference ranges and
cant improvement in seizure control, remission, adverse
individual therapeutic concentrations
effects, and quality of life measures. The direct costs
included cost to the hospital of providing the TDM service In 2008, the International League Against Epilepsy provided
and savings in cost for the hospital and patient guidelines as to how to use the terminology of reference
per seizure saved. The cost for two TDM samples ranges and the individual therapeutic concentrations [6].
per year was 60 Indian rupees, and the saving in terms
of obtained seizure control was significant, even though it 5.1.1. Reference ranges
is difficult to extract the direct savings from the results Two separate terms are recommended to define a drug con-
[77]. The study from Iraq included 132 children from 2 centration range in relation to clinical efficacy. The ‘reference
years of age with structural metabolic epileps and inves- range’ is the range of drug concentrations quoted by the
tigated the cost-effectiveness of the use of TDM retro- laboratory and specifies a lower limit, below which
spectively, according to follow-up with or without TDM. a therapeutic response is relatively unlikely to occur and an
Effectiveness was defined as >50% seizure control or sei- upper limit above which toxicity is likely to occur [2,6,80]. The
zure freedom. TDM was found cost-effective in terms of range is therefore not a ‘therapeutic range,’ which is

Figure 1. Pharmacological variability between patients and use of therapeutic drug monitoring (TDM). The figure illustrates the steps from prescription of an
antiepileptic drug to appropriate use and monitoring of serum concentrations, taking patient-specific features into account, such as adherence, pharmacokinetic,
pharmacodynamic, and pharmacogenetic variability.
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 7

associated with the best response in all patients. The thera- gabapentin, pregabalin, levetiracetam, oxcarbazepine, topir-
peutic range varies between different patients and can only be amate, and zonisamide. Emerging evidence also shows the
determined for the individual patient. Therefore, it is recom- usefulness of TDM for the third-generation AEDs such as
mended to use the term ‘individual therapeutic concentration’ brivaracetam, lacosamide, eslicarbazepine, and perampanel
[2,6,80]. and the orphan drugs, stiripentol and rufinamide. For all
It should be emphasized that because of large inter- AEDs, the indications for monitoring are the same and are
individual differences in the type of epilepsy and severity of presented in Table 2 [2,3,71,85–95].
seizures, the effective AED concentration can vary significantly
from patient to patient. Thus, patients can readily achieve 5.2.1. Other non-epilepsy comorbidities
therapeutic benefit at serum concentrations outside these AEDs are also increasingly used to treat non-epilepsy comor-
ranges. Furthermore, it is not unusual for some patients to bidities. These include, for example, gabapentin and pregaba-
have optimum seizure control at serum concentrations below lin for the management of neuropathic pain and lamotrigine,
the lower value of the reference range, while others may valproate, and carbamazepine for the management of various
require drug concentrations above the upper limit of the psychiatric conditions [92,93]. Although these AEDs show the
reference range. Therefore, while the initial treatment goal is same pharmacokinetic characteristics, they are rarely moni-
to adjust drug dosages to achieve serum concentrations tored. Interestingly, a 22-fold variability in gabapentin C/
within the reference range, it should be emphasized that it is D-ratios in a series of 120 patients with restless legs syndrome
important to treat the single patient and not the blood con- has been reported [94]. Increasingly, TDM is used to guide
centration [81]. It is thus inappropriate to adjust the dosage to antidepressant and antipsychotic treatment, and consensus
achieve blood concentrations within the reference range with- guidelines for TDM in neuropsychopharmacology from the
out taking into consideration the clinical status of the patient, German TDM task force are in place [52]. It is noteworthy
which is inappropriate. that the reference ranges for the AEDs used in psychotherapy
For many years, reference ranges have been quoted for (e.g. lamotrigine, valproate, and carbamazepine) are exactly
the first-generation AEDs (as carbamazepine, ethosuximide, the same as those used to treat epilepsy [52,96].
phenytoin, phenobarbital, and valproic acid) [82–84].
Presently, reference ranges are quoted for all the second- 5.2.2. Adherence
and third-generation AEDs [3,6,66–70]. However, because of Poor or variable adherence is a major clinical challenge and is
the considerable individual variability, the value of the the principal factor that contributes to variability in serum
lower limit of the therapeutic range has been questioned. concentrations between patients. Nonadherence is a major
Recently, a national approach to harmonize and update AED problem in the treatment of epilepsy as it compromises sei-
reference ranges between laboratories so as to improve zure control. The World Health Organization states that the
quality assurance and enhance patient management coun- use of TDM is the best way to identify and control for poor
try-wide was published by Reimers et al. [71]. adherence [97].
In children and adults with epilepsy, nonadherence has
5.1.2. Individual therapeutic concentrations been shown to be 30–40% [61,98–104]. Subtherapeutic con-
By employing the concept of ‘individual therapeutic concen- centrations, below the reference range, can also be explained
trations,’ where intra-individual comparisons of serum concen- by an inadequate low dose or by hepatic enzyme induction,
trations over time are used, TDM can be helpful for all AEDs and TDM is useful to differentiate between these, particularly
[6,80]. When an optimal effect of a drug is achieved, the if an individual reference concentration has been obtained
corresponding serum concentration will then serve as the [103]. In a study comprising 50 patients with refractory epi-
individual reference for comparison if future treatment lepsy, 40% of patients were identified as being nonadherent,
changes occur or there is a change in the clinical status of as measured by >30% change in serum concentration as
the patient. It is important to keep in mind that the basis for compared to their individual reference concentrations. Of
the application of TDM relies on a relationship between drug these patients, 66% were consuming higher doses, whereas
serum concentration and clinical effect in the individual 22% of patients consumed lower doses than prescribed (11%
patient. The optimal treatment may be best guided by identi- undefined) [102].
fying the ‘individual therapeutic concentration’ since the phar- A more recent study of 99 patients in a tertiary center reported
macokinetic variability between patients is extensive for all a lower 20% incidence of nonadherence, and this can be
AEDs [2–6,80]. explained by the fact that drug ingestion is probably better
supervised [101]. In patients admitted to hospital due to acute
seizures, serum concentrations in 40% of patients were below
5.2. Indications for therapeutic drug monitoring of AEDs
50% of their baseline steady-state concentrations [73], and 40%
The older, first-generation AEDs (carbamazepine, phenytoin, of those patients with low serum concentrations denied poor
phenobarbital, primidone, and valproic acid) exhibit exten- adherence. These data highlight that forgetting to take AEDs
sive interpatient pharmacokinetic variability and also satura- regularly has important clinical implications and should be dis-
tion kinetics for phenytoin, and therefore, TDM has been cussed with patients so that to enhance their understanding of
applied for these drugs for more than 50 years [4]. TDM is the problem and its consequence [104]. In a large Internet-based
also increasingly being used to optimize treatment with the questionnaire, it was identified that 40% of patients admitted to
various new second-generation AEDs, such as lamotrigine, forgetting drug intake sometimes/often, while 30% admitted
8 C. JOHANNESSEN LANDMARK ET AL.

that they intentionally took their AEDs differently than agreed 7. Expert opinion – future prospects
upon with their treating physician [79]. Another study was based
7.1. Expert opinion on TDM of AEDs
on questionnaires given to 175 patients and their neurologists
and showed that, in one-third of the cases, patients and their Advances in further research and application of TDM in various
treating neurologist did not agree upon the given AED(s) or AED clinical settings are promising. AED TDM is a well-established
dosing [105]. Intentional poor adherence could be due to adverse tool for the management of patients with epilepsy. Patient-
effects, concerns about polypharmacy, suitability of intake, taste specific factors contribute to extensive pharmacokinetic varia-
of tablets, etc. AED generic substitution is an emotional issue bility that may be accounted for to individualize the treatment.
whereby patients are reluctant to ingest generics because they In refractory epilepsy, a long-term TDM approach may help to
can be of different color or shape. Generics are considered bioe- solve challenges such as unsatisfactory seizure control, frequent
quivalent to brand formulations, and TDM can be useful in iden- changes in therapy, nonadherence, polypharmacy aspects, and
tifying whether any adverse effects or enhanced seizers are drug excluding pseudo-resistance due to sub-optimal use of AEDs
related [106]. There is a need for improvement in communication [73,100,104,107,108]. Also, when new treatments become avail-
between patients and health-care professionals to improve the able, pharmacokinetic challenges are evolving, and TDM is
quality of the AED treatment. a useful tool. Recent treatment options include everolimus
(tuberous sclerosis), cannabidiol (Dravet and Lennox Gastaut
syndrome), and fenfluramine (Dravet syndrome) [109]. New
5.3. Interpretation of results and impact of TDM on areas for AED TDM include other indications, such as lamotri-
clinical outcome gine in psychiatric indications and gabapentin and pregabalin
in neuropathic pain [4,9,94]. The clinical outcome may be diffi-
TDM is a useful tool for guiding the clinical management of
cult to assess also in other indications, and pharmacokinetic
patients with epilepsy. However, it is only useful if there is an
variability within and among the patients is the same regardless
appropriate indication that blood sampling is correctly underta-
of indication. Ideally, the usefulness of TDM in optimizing AED
ken and the analytical request forms are completed with the
treatment in patients with epilepsy is best ascertained by the
required clinical and sample information. Increasingly, a multi-
use of randomized controlled trial (RCT) design, and a recent
professional team comprising epileptologist, or other physician,
Cochrane review reported that they are sparse and very much
a pharmacist, a clinical pharmacologist, and an epilepsy practice
needed [110]. Indeed, there are two previous RCTs comparing
nurse is in place and can result in a robust effort to provide optimal
treatment outcome guided with the use of TDM [111,112].
patient care. While the reference range is useful in the first
These studies were not able to provide sufficient evidence for
instance to guide treatment, it is important that the individual
the usefulness of routine monitoring of AEDs in the general
therapeutic concentration is identified as quickly as possible.
epilepsy population. This does not argue against the value of
TDM in special situations and clinical indications. The nature of
RCTs to study the usefulness of TDM is not possible because
6. Conclusion
patient-related factors and use of TDM on clinical grounds
Table 1 highlights the principal indications for AED TDM, make blinding and randomization difficult. An unsuccessful
while Table 2 highlights the useful pharmacokinetic characteris- example of this is a recent prospective RCT during pregnancy
tics that are needed so as to aid implementation and interpreta- with or without TDM. No difference between the groups on
tion of TDM data. Because TDM provides a pragmatic approach seizure control or outcome in pregnant women could be
to epilepsy care with AEDs, AEDs are among the most common demonstrated due to several limiting factors [113].
medications for which TDM is undertaken. Indeed, AED TDM
enables bespoke dose adjustments based on actual drug con-
centration measurements so as to optimize treatment outcome.
7.2. Future prospects of precision medicine in epilepsy
It is important to understand the concept of the reference range
because for the majority of patients, if their serum concentrations Precision medicine entails tailored or personalized treatment.
are within these ranges, they will exhibit optimum clinical Going forward, we can expect new and improved analytical
response. Some patients, however, may require concentrations procedures that will enable a more sensitive analysis to be
outside the reference ranges consequent to individual variation performed so that low concentrations of drugs and metabo-
in seizure severity and seizure type. Equally, it is important to lites can be more readily measured and more rapid and accu-
understand and to apply the concept of the ‘individual thera- rate results to be made available quickly. Genotyping of CYP
peutic concentration’ which is specific for an individual patient isoenzymes such as CYP2C9 and 2C19, and UGT isoenzymes, is
and reflects his/her optimum treatment regimen. likely to become available for a specific use. These tests may
Serum or plasma is the matrix of choice for AED TDM. allow avoidance of overdosing due to reduced activity of
However, with the introduction of sensitive analytical instru- metabolic pathways, and thus, patient optimum treatment
mentation, many AEDs can be readily monitored in saliva or will be achieved earlier. Pharmacogenomic approaches
dried blood spots. Increasingly a multi-professional team com- include screening of all gene variants and thus reveal more
prising epileptologist, or other physician, a pharmacist, causes of e.g. pharmacoresistance [56]. An example is the
a clinical pharmacologist, and an epilepsy practice nurse is in identification of SCN1A mutations in Dravet syndrome,
place to interpret TDM data, and this can result in a robust where the voltage-gated sodium channel has a loss of func-
effort to provide optimal patient care. tion. Thus, the introduction of drugs that work via an action on
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 9

SVN1A would be ineffective and consequently may worsen treatment by implementation of therapeutic drug monitoring.
seizures and inadvertently result in refractory epilepsy. Epileptic Disord. 2016;18(4):367–383.
5. Johannessen SI, Battino D, Berry DJ, et al. Therapeutic drug mon-
As a supplement to TDM as part of precision medicine,
itoring of the newer antiepileptic drugs. Ther Drug Monit.
measurement of more pharmacogenetic and biochemical mar- 2003;25:347–363.
kers of toxicity may lead the way forward to avoid toxicity at 6. Patsalos PN, Berry DJ, Bourgeois BF, et al. Antiepileptic drugs – best practice
early stages in the treatment. The best example is the HLA- guidelines for therapeutic drug monitoring: a position paper by the sub-
1502 mutations, frequently occurring in South-East Asians, and commission on therapeutic drug monitoring, ILAE commission on therapeu-
tic strategies. Epilepsia. 2008;49:1239–1276.
with increased risk of Stevens–Johnson syndrome consequent
•• International guidelines for AED TDM.
to carbamazepine and phenytoin treatment. In clinical prac- 7. Patsalos PN, Zugman M, Lake C, et al. Serum protein binding of 25
tice, pharmacogenetic markers of both HLA and CYP muta- antiepileptic drugs in a routine clinical setting: a comparison of free
tions could be tested as a panel to assess risk factors [114– non-protein-bound concentrations. Epilepsia. 2017;58
116]. Another new approach is to use an endogenous biomar- (7):1234–1243.
•• The only study describing details on protein binding in
ker, 4β-hydroxycholesterol, for CYP3A-activity to evaluate the
a clinical setting, which will be important for further analytical
potency of enzyme-inducing capacity [38]. Another approach progress and precise measurements of the pharmacologically
that will possibly be applied and developed in the years to active component of many AEDs.
come is to use population pharmacokinetic modeling so as to 8. Patsalos PN, Berry DJ. Therapeutic drug monitoring of antiepi-
aim at an initial target dosage regime, which can subsequently leptic drugs by use of saliva. Ther Drug Monit. 2013;35:4–29.
9. Baftiu A, Feet SA, Larsson PG, et al. Utilization of antiepileptic drugs
be followed by TDM in the individual patient [2]. Such model-
in the elderly vs younger patients with epilepsy and psychiatric
ing allows for dosage adjustments to be undertaken before comorbidity. Epilepsy Res. 2018;139:35–42.
reaching steady state and before the measurement of serum 10. Chen Z, Brodie MJ, Liew D, et al. Treatment outcomes with newly
concentrations. This could help clinicians to more rapidly diagnosed epilepsy treated with established and new antiepileptic
establish an effective dose. Also, such software includes algo- drugs: a 30-year longitudinal cohort study. JAMA Neurol. 2018;75
(3):279–286.
rithms so as to allow the determination of pharmacokinetic
•• Large longitudinal study confirming that one-third of patients
variability, drug interactions, and pharmacogenetic factors to continue to be refractory to AED treatment.
provide a patient-oriented service [2]. 11. Kwan P, Brodie MJ. Early identification of refractory epilepsy. N Engl
J Med. 2000;342:314–319.
•• One of the main references to demonstrate that one-third of
Funding patients are treatment resistant, reconfirmed by Chen et al.,
2018.
This paper was not funded. 12. Glauser T, Ben-Menachem E, Bourgeois B, et al. Updated ILAE
evidence review of antiepileptic drug efficacy and effectiveness
as initial monotherapy for epileptic seizures and syndromes.
Declaration of interest Epilepsia. 2013;54(3):551–563.
13. Nunes VD, Sawyer L, Neilson J, et al. Diagnosis and management of
The authors have no relevant affiliations or financial involvement with any
the epilepsies in adults and children: summary of updated NICE
organization or entity with a financial interest in or financial conflict with
guidance. BMJ. 2012;26:e344:281.
the subject matter or materials discussed in the manuscript. This includes
14. Hadjiloizou SM, Bourgeois BF. Antiepileptic drug treatment in
employment, consultancies, honoraria, stock ownership or options, expert
children. Expert Rev Neurother. 2007;7:179–193.
testimony, grants or patents received or pending, or royalties.
15. Italiano D, Perucca E. Clinical pharmacokinetics of new-generation
antiepileptic drugs at the extremes of age: an update. Clin
Pharmacokinet. 2013;8:627–645.
Reviewer disclosures 16. Verotti A, Iapadre G, Di Donato G, et al. Pharmacokinetic considera-
Peer reviewers on this manuscript have no relevant financial or other tions for anti-epileptic drugs in children. Exp Opin Drug Metab
relationships to disclose. Toxicol. 2019;15(3):199–211.
17. Tomson T, Landmark CJ, Battino D. Antiepileptic drug treatment in
pregnancy: changes in drug disposition and their clinical
References implications. Epilepsia. 2013;54:405–414.
18. Patel SI, Pennell PB. Management of epilepsy during pregnancy: an
Papers of special note have been highlighted as either of interest (•) or of update. Ther Adv Neurol Disord. 2016;9(2):118–129.
considerable interest (••) to readers. 19. Tomson T, Battino D. Pharmacokinetics and therapeutic drug mon-
1. Patsalos PN, St Louis EK. The epilepsy prescriber’s guide to anti- itoring of newer antiepileptic drugs during pregnancy and the
epileptic drugs. 3rd ed. Cambridge: Cambridge University Press; puerperium. Clin Pharmacokinet. 2007a;46:209–219.
2018. 20. Petrenaite V, Sabers A, Hansen-Schwartz J. Individual changes in
2. Patsalos PN, Spencer EP, Berry DJ. Therapeutic drug monitoring of lamotrigine plasma concentrations during pregnancy. Epilepsy Res.
antiepileptic drugs in epilepsy: a 2018 update. Ther Drug Monit. 2005;65:185–188.
2018;40(5):526–548. 21. Pennell PB, Peng L, Newport DJ, et al. Lamotrigine in pregnancy.
• The most recent update on TDM in epilepsy with details on the Clearance, therapeutic drug monitoring, and seizure frequency.
various drugs. Neurology. 2008;70:2130–2136.
3. Johannessen Landmark C, Johannessen SI, Tomson T. Host factors 22. Tomson T, Palm R, Källén K, et al. Pharmacokinetics of levetirace-
affecting antiepileptic drug delivery – pharmacokinetic variability. tam during pregnancy, delivery, in the neonatal period, and
Adv Drug Deliv Rev. 2012a;64:896–910. lactation. Epilepsia. 2007b;48:1111–1116.
• Thorough update on pharmacokinetic variability of AEDs. 23. Mazzucchelli I, Onat FY, Özkara C, et al. Changes in the disposition
4. Johannessen Landmark C, Johannessen SI, Tomson T. Dosing stra- of oxcarbazepine and its metabolites during pregnancy and the
tegies of antiepileptic drugs – from one dose to individualisation of puerperium. Epilepsia. 2006;47(3):504–509.
10 C. JOHANNESSEN LANDMARK ET AL.

24. Öhman I, Vitols S, Luef G, et al. Topiramate kinetics during delivery, 44. Burns ML, Baftiu A, Opdal MS, et al. Therapeutic drug monitoring of
in the neonatal period, and lactation: preliminary observations. clobazam and its metabolite-Impact of age and comedication on
Epilepsia. 2002;43:1157–1160. pharmacokinetic variability. Ther Drug Monit. 2016;38(3):350–357.
25. Westin AA, Nakken KO, Johannessen SI, et al. Serum concentration/ 45. Karouni M, Henning O, Larsson PG, et al. Psychiatric comorbidity in
dose ratio of topiramate during pregnancy. Epilepsia. patients with refractory epilepsy. Epi Behav. 2013;29:77–81.
2009;50:480–485. 46. Spina E, Pisani F, de Leon J. Clinically significant pharmacokinetic
26. Reimers A, Helde G, Becser Andersen N, et al. Zonisamide serum drug interactions of antiepileptic drugs with new antidepressants
concentrations during pregnancy. Epilepsy Res. 2018a;144:25–29. and new antipsychotics. Pharmacol Res. 2016;106:72–86.
27. Öhman I, Vitols S, Tomson T. Pharmacokinetics of gabapentin 47. Asconapé JJ. Pharmacokinetic considerations with the use of anti-
during delivery, in the neonatal period, and lactation: does a fetal epileptic drugs in patients with HIV and organ transplants. Curr
accumulation occur during pregnancy? Epilepsia. Neurol Neurosci Rep. 2018;18:89.
2005;46:1621–1624. 48. Sabers A, Öhman I, Christensen J, et al. Oral contraceptives reduce
28. Gerard EE, Meador KJ. An update on maternal use of antiepileptic lamotrigine plasma levels. Neurology. 2003;61:570–571.
medications in pregnancy and neurodevelopment outcomes. 49. Reimers A, Helde G, Brodtkorb E. Ethinyl estradiol, not progestogens,
J Pediatr Genet. 2015;4(2):94–110. reduces lamotrigine serum concentrations. Epilepsia.
29. EMA. Visited March 28th, 2019. https://www.ema.europa.eu/en/ 2005;46:1414–1417.
medicines/human/referrals/valproate-related-substances. 50. Galgani A, Palleria C, Iannone LF, et al. Pharmacokinetic interac-
30. Tomson T, Battino D, Bonizzoni E, et al. Comparative risk of major tions of clinical interest between direct oral anticoagulants and
congenital malformations with eight different antiepileptic drugs: antiepileptic drugs. Front Neurol. 2018;9:1067.
a prospective cohort study of the EURAP registry. Lancet Neurol. 51. Löscher W, Klotz U, Zimprich F, et al. The clinical impact of pharma-
2018;17(6):530–538. cogenetics on the treatment of epilepsy. Epilepsia. 2009;50(1):1–23.
•• Results from a large international registry on pregnancy 52. Hiemke C, Bergemann N, Clement HW, et al. Consensus guidelines
outcomes. for therapeutic drug monitoring in neuropsychopharmacology:
31. Johannessen Landmark C, Burns ML, Baftiu A, et al. update 2017. Pharmacopsychiatry. 2018;51:9–62.
Pharmacokinetic variability of valproate in women of 53. Bozina N, Sporis IS, Bozina T, et al. Pharmacogenomics and the
childbearing. Epilepsia. 2017;58(10):e142–e146. treatment of epilepsy: what do we know?. Pharmacogenomics.
32. Johannessen Landmark C, Farmen AH, Burns ML, et al. 2019;20(15):1093–1101.
Pharmacokinetic variability of valproate during pregnancy – implica- • Recent update on the role of pharmacogenomics in epilepsy.
tions for the use of therapeutic drug monitoring. Epilepsy Res. 54. Smith RL, Haslemo T, Refsum H, et al. Impact of age, gender and
2018;141:31–37. CYP2C9/2C19 genotypes on dose-adjusted steady-state serum con-
33. Johannessen SI, Johannessen Landmark C. Antiepileptic drug interac- centrations of valproic acid – a large-scale study based on natur-
tions – basic principles and clinical implications. Curr Neuropharm. alistic therapeutic drug monitoring data. Eur J Clin Pharmacol.
2010;8:254–267. 2016;72(9):1099–1104.
34. Johannessen Landmark C, Patsalos PN. Drug interactions involving 55. Smith RL, Haslemo T, Chan HF, et al. Clinically relevant effect of
the new second- and third-generation antiepileptic drugs. Expert UGT1A4*3 on lamotrigine serum concentration is restricted to
Rev Neurother. 2010;10:119–140. postmenopausal women – a study matching therapeutic drug
35. Patsalos PN. Drug interactions with the newer antiepileptic drugs monitoring and genotype data from 534 patients. Ther Drug
(AEDs)-part 1: pharmacokinetic and pharmacodynamic interactions Monit. 2018;40(5):567–571.
between AEDs. Clin Pharmacokinet. 2013a;52:927–966. 56. Tang F, Hartz AMS, Bauer B. Drug-resistant epilepsy: multiple
36. Patsalos PN. Drug interactions with the newer antiepileptic drugs hypotheses, few answers. Front Neurol. 2017;8:301. eCollection
(AEDs)-part 2: pharmacokinetic and pharmacodynamic interactions 2017.
between AEDs and drugs used to treat non-epilepsy disorders. Clin • Recent update discussing various hypothesis for drug
Pharmacokinet. 2013b;52:1045–1061. resistance.
37. Patsalos PN. Antiepileptic drug interactions: a clinical guide. 3rd ed. 57. Mirza N, Vaseiva O, Appleton R. An integrative in silico system for
Switzerland: Springer; 2016. predicting dysregulated genes in the human epileptic focus: appli-
•• A useful and concise guide that describes all known interac- cation to SLC transporters. Epilepsia. 2016;57:1467–1474.
tions involving AEDs. 58. Franco V, Baruffi K, Fransesca Crema TM, et al. Determination of
38. Hole K, Wollmann BM, Nguyen C, et al. Comparison of CYP3A4- perampanel in dried blood spots; applicability to therapeutic
inducing capacity of enzyme-inducing antiepileptic drugs using drug monitoring. Ther Drug Monit. 2019 Jul 29. Epub ahead of
4β-hydroxycholesterol as biomarker. Ther Drug Monit. 2018;40 print.
(4):463–468. 59. Velghe S, Deprez S, Stove CP. Fully automated therapeutic drug
• New approach to estimate the enzyme-inducing potency of monitoring of antiepileptic drugs making use of dried blood spots.
inducing AEDs. J Chromatogr A. 2019;1601:95–103.
39. Bertilsson L, Höjer B, Tybring G, et al. Autoinduction of carbamaze- 60. Wilhelm AJ, den Burger JC, Swart EL. Therapeutic drug monitoring
pine metabolism in children examined by a stable isotope by dried blood spot: progress to date and future directions. Clin
technique. Clin Pharmacol Ther. 1980;27:83–88. Pharmacokinet. 2014;53(11):961–973.
40. Kudriakova TB, Sirota LA, Rozova GI, et al. Autoinduction and 61. Shah NM, Hawwa AF, Millership JS, et al. Adherence to antiepileptic
steady-state pharmacokinetics of carbamazepine and its major medicines in children: a multiple-methods assessment involving
metabolites. Br J Clin Pharmacol. 1992;33:611–615. dried blood spot sampling. Epilepsia. 2013;54:1020–1027.
41. Johannessen Landmark C, Baftiu A, Tysse I, et al. Pharmacokinetic 62. Linder C, Neideman M, Wide K, et al. Dried blood spot
variability of four newer antiepileptic drugs, lamotrigine, levetira- self-sampling by guardians of children with epilepsy is feasible;
cetam, oxcarbazepine and topiramate-a comparison of the impact comparison with plasma for multiple antiepileptic drugs. Ther
of age and comedication. Ther Drug Monit. 2012b;34(4):440–445. Drug Monit. 2019;41(4):509–518.
42. Koristkova B, Grundmann M, Brozmanova H, et al. Lamotrigine 63. Linder C, Wide K, Walander M, et al. Comparison between dried
drug interactions in combination therapy and the influence of blood spot and plasma sampling for therapeutic drug monitoring
therapeutic drug monitoring on clinical outcomes in paediatric of antiepileptic drugs in children with epilepsy: a step towards
patients. Basic Clin Pharmacol Toxicol. 2019;125(1):26–33. home sampling. Clin Biochem. 2017;50(7–8):418–424.
43. De Leon J, Spina E, Diaz FJ. Clobazam therapeutic drug monitoring: 64. D’Urso A, Cangemi G, Barco S, et al. LC-MS/MS-based quantification
a comprehensive review of the literature with proposals to improve of 9 antiepileptic drugs from a dried sample spot device. Ther Drug
future studies. Ther Drug Monit. 2013;35(1):30–47. Monit. 2019;41(3):331–339.
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 11

65. D’Urso A, Rudge J, Patsalos PN, et al. Volumetric absorptive micro- 86. Svendsen T, Brodtkorb E, Reimers A, et al. Pharmacokinetic
sampling: a new sampling tool. Ther Drug Monit. 2019;41 variability, efficacy and tolerability of eslicarbazepine acetate –
(5):681–692. a national approach to the evaluation of therapeutic drug mon-
66. Johannessen SI, Gerna M, Bakke J, et al. CSF concentrations and itoring data and clinical outcome. Epilepsy Res.
serum protein binding of carbamazepine and 2017a;129:125–131.
carbamazepine-10,11-epoxide in epileptic patients. Br J Clin 87. Svendsen T, Brodtkorb E, Baftiu A, et al. Therapeutic drug monitor-
Pharmacol. 1976;3:575–582. ing of lacosamide in Norway: focus on pharmacokinetic variability,
67. Bertilsson L, Tomson T. Clinical pharmacokinetics and pharmacolo- efficacy and tolerability. Neurochem Res. 2017b;42(7):2077–2083.
gical effects of carbamazepine and carbamazepine-10,11-epoxide. 88. Steinhoff BJ, Hübers E, Kurth C, et al. Plasma concentration and clinical
An Update Clin Pharmacokinet. 1986;11:177–198. effects of perampanel – the Kork experience. Seizure. 2019;28:18–22.
68. Pisani F, Caputo M, Fazio A, et al. Interaction of carbamazepine-10- 89. Burns ML, Nikanorova M, Baftiu A, et al. Pharmacokinetic varia-
11-epoxide, an active metabolite of carbamazepine, with valproate: bility and clinical use of lacosamide in children and adolescents
a pharmacokinetic study. Epilepsia. 1990;31:339–342. in Denmark and Norway. Ther Drug Monit. 2019a;41
69. Burianová I, Borecka K. Routine therapeutic monitoring of the (3):340–347.
active metabolite of carbamazepine: is it really necessary? Clin 90. Contin M, Albani F, Riva R, et al. Lacosamide therapeutic monitor-
Biochem. 2015;48:866–869. ing in patients with epilepsy: effect of concomitant antiepileptic
70. Stockis A, Sargentini-Maier ML, Brodie MJ. Pharmacokinetic inter- drugs. Ther Drug Monit. 2013;35(6):849–852.
action of brivaracetam on carbamazepine in adult patients with 91. May TW, Helmer R, Bien CG, et al. Influence of dose and antiepi-
epilepsy, with and without valproate co-administration. Epilepsy leptic comedication on lacosamide serum concentrations in
Res. 2016;128:163–168. patients with epilepsy of different ages. Ther Drug Monit. 2018;40
71. Reimers A, Andsnes Berg J, Larsen Burns M, et al. Reference ranges (5):620–627.
for antiepileptic drugs revisited: a practical approach to establish 92. Johannessen Landmark C, Larsson PG, Rytter E, et al. Antiepileptic
national guidelines. Drug Des Devel Ther. 2018b;12:271–280. drugs in epilepsy and other disorders: a population-based study of
72. Specht U, Elsner H, May TW, et al. Postictal serum levels of anti- prescriptions. Epilepsy Res. 2009;87:31–39.
epileptic drugs for detection of noncompliance. Epilepsy Behav. 93. Baftiu A, Johannessen Landmark C, Rudberg Rusten I, et al. Safety
2003;4(5):487–495. aspects of utilization of antiepileptic drugs in epilepsy vs.
73. Samsonsen C, Reimers A, Bråthen G, et al. Nonadherence to treat- non-epilepsy disorders. Eur J Clin Pharmacol. 2016;72
ment causing acute hospitalizations in people with epilepsy: an (10):1245–1254.
observational, prospective study. Epilepsia. 2014;55(11):e125–128. 94. Burns ML, Kinge E, Stokke Opdahl M, et al. Therapeutic drug
• Prospective study to identify poor adherence by using TDM. monitoring of gabapentin in various indications. Acta Neurol
74. Touw DJ, Neef C, Thomson AH, et al. Cost-effectiveness of thera- Scand. 2019b;130(5):446–454.
peutic drug monitoring: a systematic review. Ther Drug Monit. 95. Conway JM, Collins JF, Macias FM, et al. Factors in variability of
2005;27:10–17. serial gabapentin concentrations in elderly patients with epilepsy.
75. Eadie MJ. Plasma antiepileptic drug monitoring in a neurological Pharmacotherapy. 2017;37(10):1197–1203.
practice: a 25-year experience. Ther Drug Monit. 1994;16:458–468. 96. Bengtsson F. Therapeutic drug monitoring of psychotropic drugs.
76. Eadie MJ. The role of therapeutic drug monitoring in improving the TDM “nouveau”. Ther Drug Monit. 2004;26:145–151.
cost effectiveness of anticonvulsant therapy. Clin Pharmacokinet. 97. World Health Organization. Adherence to long term therapies –
1995;29:29–35. evidence for action. World Health Organization 2003. Available
77. Rane CT, Dalvi SS, Gogtay NJ, et al. A pharmacoeconomic analysis from: http://apps.who.int/iris/bitstream/handle/10665/42682/
of the impact of therapeutic drug monitoring in adult patients with 9241545992.pdf
generalized tonic-clonic epilepsy. Br J Clin Pharmacol. 98. Davis KL, Candrilli SD, Edin HM. Prevalence and cost of nonadher-
2001;52:193–195. ence with antiepileptic drugs in an adult managed care population.
78. Salih MR, Bahari MB, Shafie AA, et al. Cost-effectiveness analysis Epilepsia. 2008;49:446–454.
for the use of serum antiepileptic drug level monitoring in chil- 99. Henning O, Johannessen Landmark C, Nakken KO, et al.
dren with structural-metabolic epilepsy. Epilepsy Res. Nonadherence to treatment regimens in epilepsy from the
2013;104:151–157. patients’ perspective and predisposing factors: differences
79. Hussain SA, Ortendahl JD, Bentley TGK, et al. The economical between intentional and non-intentional lack of adherence.
burden of caregiving in epilepsy: an estimate based on Epilepsia. 2019a;60(5):e58–e62.
a survey of US caregivers. Epilepsia. 2020 Jan 17. Epub ahead 100. Johannessen Landmark C, Fløgstad I, Syvertsen M, et al. Treatment
of print. with AEDs and challenges for patients with JME. Epi Behav.
80. Perucca E. Is there a role for therapeutic drug monitoring of new 2019;98:110–116.
anticonvulsants? Clin Pharmacokinet. 2000;38:191–204. 101. Henning O, Lossius MI, Lima MH, et al. Refractory epilepsy and
• First paper to propose the concept of individual therapeutic nonadherence to drug treatment. Epilepsia Open.
concentrations. 2019b;4:618–623.
81. Woo E, Chan YM, Yu YL, et al. If a well-stabilized epileptic patient 102. Carpentier N, Jonas J, Frismand S, et al. Direct evidence of non-
has a subtherapeutic antiepileptic drug level, should the dose be adherence to antiepileptic medication in refractory focal epilepsy.
increased? A randomized prospective study. Epilepsia. Epilepsia. 2013;54(1):e20–e23.
1988;29:129–139. 103. Lunardi M, Lin K, Walz R, et al. Single antiepileptic drug levels do
82. Buchthal F, Svensmark O, Schiller PJ. Clinical and electroencephalo- not predict adherence and nonadherence. Acta Neurol Scand.
graphic correlations with serum levels of diphenylhydantoin. Arch 2019;139(2):199–203.
Neurol. 1960;2:624–630. 104. Johannessen Landmark C, Fløgstad I, Baftiu A, et al. Long term
83. Schmidt D, Haenel F. Therapeutic plasma levels of phenytoin, therapeutic drug monitoring of AEDs in patients with JME. Epilepsy
phenobarbital, and carbamazepine: individual variation in relation Res. 2019;155:106148.
to seizure frequency and type. Neurology. 1984;34:1252–1255. 105. Mevaag M, Henning O, Baftiu A, et al. Discrepancies between
84. Schmidt D, Einicke I, Haenel FT. The influence of seizure type on physicians’ prescriptions and patients’ use of antiepileptic drugs.
the efficacy of plasma concentrations of phenytoin, phenobarbital, Acta Neurol Scand. 2017;135:80–87.
and carbamazepine. Arch Neurol. 1986;43:263–265. 106. Holtkamp M, Theodore WH. Generic antiepileptic drugs – safe or
85. Gidal BE, Ferry J, Majid O, et al. Concentration-effect relationships harmful in patients with epilepsy? Epilepsia. 2018;59(7):1273–1281.
with perampanel in patients with pharmacoresistant partial-onset 107. Kwan P, Arzimanoglou A, Berg AT, et al. Definition of drug resistant
seizures. Epilepsia. 2013;54:1490–1497. epilepsy: consensus proposal by the ad hoc task force of the ILAE
12 C. JOHANNESSEN LANDMARK ET AL.

commission on therapeutic strategies. Epilepsia. monitoring in patients with newly diagnosed epilepsy. Epilepsia.
2010;51:1069–1077. 2000;41:222–230.
• Consensus document on treatment-resistant epilepsy. 113. Thangaratinam S, Marlin N, Newton S, et al. AntiEpileptic drug
108. Hao X, Goldberg D, Kelly K, et al. Uncontrolled epilepsy is not monitoring in PREgnancy (EMPiRE): a double-blind randomized
necessarily the same as drug-resistant epilepsy: differences trial on effectiveness and acceptability of monitoring strategies.
between populations with newly diagnosed epilepsy and chronic Health Technol Assess. 2018;22(23):1–152.
epilepsy. Epilepsy Behav. 2013;29(1):4–6. 114. Chen Z, Liew D, Kwan P. Real-world efficiency of pharmacogenetic
109. Bialer M, Johannessen SI, Koepp MJ, et al. Progress report on new screening for carbamazepine-induced severe cutaneous adverse
antiepileptic drugs: a summary of the fourteenth Eilat conference on reactions. PLoS One. 2014;9(5):e96990.
new antiepileptic drugs and devices (EILAT XiV) II. Drugs in more •• Evidence for the efficiency of screening for mutations in order
advanced clinical development. Epilepsia. 2018;59(10):1842–1866. to avoid carbamazepine-induced toxicity and thus avoiding
• Update on the development of AEDs that are undergoing clinical exposure to vulnerable patients.
studies. 115. Tassaneeyakul W, Prabmeechai N, Sukasem C, et al. Associations
110. Tomson T, Dahl M, Kimland E. Therapeutic monitoring of antiepileptic between HLA class I and cytochrome P450 2C9 genetic polymorphisms
drugs for epilepsy. Cochrane Database Syst Rev. 2007c;1:CD002216. and phenytoin-related severe cutaneous adverse reactions in a Thai
111. Fröscher W, Eichelbaum M, Gugler R, et al. A prospective rando- population. Pharmacogenet Genomics. 2016;26(5):225–234.
mised trial on the effect of monitoring plasma anticonvulsant levels 116. Chouchi M, Kaabachi W, Tizaoui K, et al. The HLA-B*15:02 poly-
in epilepsy. J Neurol. 1981;224:193–201. morphism and Tegretol®-induced serious cutaneous reactions in
112. Jannuzzi G, Cian P, Fattore C, et al. A multicenter randomized epilepsy: an updated systematic review and meta-analysis. Rev
controlled trial on the clinical impact of therapeutic drug Neurol (Paris). 2018;174(5):278–291.

View publication stats

You might also like