You are on page 1of 15

Pharmacological Research 106 (2016) 72–86

Contents lists available at ScienceDirect

Pharmacological Research
journal homepage: www.elsevier.com/locate/yphrs

Review

Clinically significant pharmacokinetic drug interactions of


antiepileptic drugs with new antidepressants and new antipsychotics
Edoardo Spina a,∗ , Francesco Pisani a , Jose de Leon b,c,d
a
Department of Clinical and Experimental Medicine, University of Messina, Italy
b
University of Kentucky Mental Health Research Center at Eastern State Hospital, Lexington, KY, United States
c
Psychiatry and Neurosciences Research Group (CTS-549), Institute of Neurosciences, University of Granada, Granada, Spain
d
Biomedical Research Centre in Mental Health Net (CIBERSAM), Santiago Apostol Hospital, University of the Basque Country, Vitoria, Spain

a r t i c l e i n f o a b s t r a c t

Article history: Antiepileptic drugs (AEDs) are frequently co-prescribed with new antidepressants (ADs) or new antipsy-
Received 21 January 2016 chotics (APs). A PubMed search with no time limit was used to update the review of the clinically
Received in revised form 8 February 2016 significant pharmacokinetic (PK) drug interactions DIs (DIs) between AEDs with new ADs and APs. Our
Accepted 11 February 2016
best interpretation of what to expect regarding dosing changes in the average patient after combining
Available online 16 February 2016
AEDs with new ADs or new APs is summarized on updated tables that integrate the information on
in vitro metabolism studies, therapeutic drug monitoring (TDM) studies, case report/series and prospec-
Keywords:
tive studies. There will be a need to periodically update these dose correction factors as new knowledge
Antiepileptics
Antidepressants becomes available. These tables will provide some orientation to clinicians with no TDM access and
Antipsychotics may also encourage clinicians to further study TDM. The clinical relevance of the inductive proper-
Drug interactions ties of carbamazepine, phenytoin, phenobarbital and primidone on new ADs and new APs and the
Pharmacodynamics inhibitory properties of valproic acid and some new ADs, are relatively well understood. On the other
Pharmacokinetics hand, PK DI studies combining new AEDs with weak inductive properties (particularly oxcarbazepine
doses ≥ 1200 mg/day), topiramate doses ≥ 400 mg/day, clobazam, eslicarbazepine, and rufinamide), with
new ADs and new APs are needed. Valproic acid may be 1) an inhibitor and/or inducer of clozapine and
olanzapine with potential for clinically relevant DIs, 2) an inhibitor of paliperidone, and 3) a weak inducer
of aripiprazole. Fluoxetine and fluvoxamine are relevant inhibitors of phenytoin and valproic acid and
possibly of clobazam, lacosamide, phenobarbital, or primidone.
© 2016 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
2. Basic mechanisms of DIs between AEDs and new psychotropic agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
2.1. PK DIs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
2.2. PD DIs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
3. DIs between AEDs and new ADs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
3.1. Effect of AEDs on the PKs of new ADs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
3.1.1. Enzyme-inducing AEDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
3.1.2. Valproic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
3.1.3. Newer AEDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
3.2. Effect of new ADs on the PKs of AEDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
3.2.1. SSRIs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
3.2.2. SNRIs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78

∗ Corresponding author at: Department of Clinical and Experimental Medicine,


University of Messina, Policlinico Universitario, Via Consolare Valeria, 98125
Messina, Italy.
E-mail address: espina@unime.it (E. Spina).

http://dx.doi.org/10.1016/j.phrs.2016.02.014
1043-6618/© 2016 Elsevier Ltd. All rights reserved.
E. Spina et al. / Pharmacological Research 106 (2016) 72–86 73

3.2.3. Other newer antidepressants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78


4. DIs between AEDs and new APs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
4.1. Effect of AEDs on the PKs of new APs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
4.1.1. Carbamazepine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
4.1.2. Phenobarbital/phenytoin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
4.1.3. Valproic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 80
4.1.4. Lamotrigine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 80
4.1.5. Oxcarbazepine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81
4.1.6. Topiramate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
4.1.7. Other newer AEDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
4.2. Effect of new APs on the PKs of AEDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
5. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84

1. Introduction sive disorder, psychosis and behavioral disorders in dementia,


psychosis associated with Parkinson’s disease, resistant obsessive-
When polypharmacotherapy is used, there is a potential risk of compulsive disorder, aggressive behavior and irritability in autism
drug interactions (DIs) [1]. A clinically relevant DI occurs when the spectrum disorders [5].
efficacy or safety of a drug is altered by the concomitant administra- Considering the frequent co-prescription of AEDs with new psy-
tion of another medication. In a few cases DIs may prove beneficial, chotropic agents, it is essential for clinicians to be aware of the
resulting in an increased efficacy or reduced risk of adverse drug potential DIs between these compounds. In recent years, a number
reactions, and therefore certain drug combinations may be used of comprehensive reviews of clinically relevant pharmacokinetic
advantageously in clinical practice. However, more often, DIs are DIs involving AEDs [2,6–11], new ADs [12,13], new APs [14,15],
harmful, leading to diminished efficacy or increased toxicity of one or combinations [3,16–20] have been published. The purpose of
or more of the administered medications. this article is to provide an updated review of clinically significant
DIs represent a frequent complication associated with the use of pharmacokinetic DIs between AEDs and new ADs and APs.
antiepileptic drugs (AEDs) [2]. AEDs are generally administered for Articles for this review were obtained from a PubMed search
prolonged periods, often for a lifetime, increasing the probability with no time limit. Searches were conducted for each of the AEDs,
of co-medication with other AEDs or drugs used for the manage- new ADs or new APs. Only articles published in peer-reviewed
ment of associated disorders. In this respect, AEDs are increasingly journals were included, while meeting abstracts were excluded.
prescribed in combination with psychotropic agents for a variety of Information was also obtained from the individual product inserts
reasons [3]. Firstly, there is a relatively high incidence of psychiatric of each AED, new AD or new AP. Additional DI information was also
disorders in patients with epilepsy. Secondly, a number of AEDs, obtained from citations of the articles that were retrieved during
notably carbamazepine, valproic acid and lamotrigine, are increas- our search, and these were also included in our review. This search
ingly used in the management of psychiatric disorders, generally was beyond the articles previously found and listed in the authors’
as mood stabilizers, and may be co-administered with psychoac- published literature reviews and DI studies.
tive medications. Thirdly, AEDs are extensively prescribed for the
management of various non-epileptic conditions (i.e., neuropathic 2. Basic mechanisms of DIs between AEDs and new
pain, migraine), further enhancing the possibility of combination psychotropic agents
therapy.
In recent years, new psychotropic drugs, in particular antide- Based on their mechanisms, DIs can be classified as either phar-
pressants (ADs) and antipsychotics (APs), have been introduced macokinetic (PK) or pharmacodynamic (PD).
into clinical practice. In addition to older or traditional ADs,
such as tricyclic antidepressants (TCAs) and monoamine oxi- 2.1. PK DIs
dase inhibitors (MAOIs), newer ADs including selective serotonin
reuptake inhibitors (SSRIs), serotonin and noradrenaline reuptake PK DIs consist of changes in the absorption, distribution,
inhibitors (SNRIs) and other ADs with varying mechanisms of action metabolism or excretion of a drug and/or its metabolite(s) after the
are currently available [4]. Some of these agents, notably SSRIs, addition of another drug. These DIs are associated with a modifica-
have progressively replaced older compounds for the treatment of tion in plasma concentration of either the drug or its metabolite(s)
depressive disorders, mainly because of their improved tolerability and usually are easily verified by therapeutic drug monitoring
and safety profile. New ADs are also widely used for the treatment of (TDM). PK parameters of AEDs, newer ADs and newer APs are sum-
other psychiatric conditions including anxiety disorders, obsessive- marized in Tables 1– , respectively.
compulsive disorder, eating disorders, and various forms of chronic The majority of clinically important PK DIs between AEDs and
pain such as diabetic neuropathic pain and fibromyalgia [4]. Con- new psychotropic agents occur at a metabolic level and usu-
cerning APs, over the past two decades, second-generation APs ally involve the hepatic cytochrome P450 (CYP) system and, to
(amisulpride, aripiprazole, asenapine, brexpiprazole, cariprazine, a lesser extent, the uridine diphosphate glucuronosyltransferase
clozapine, iloperidone, lurasidone, olanzapine, paliperidone, que- (UGT) system [21]. In recent years, the in vitro characterization of
tiapine,risperidone, and ziprasidone) have become the drugs of the major drug-metabolizing enzymes with identification of sub-
choice for the management of psychotic disorders due to better strates, inhibitors and inducers of different CYP isoforms has greatly
safety profiles for reversible extrapyramidal symptoms and tar- improved the prediction of metabolic DIs, providing an invaluable
dive dyskinesia as compared to first-generation APs [5]. Moreover, resource in helping to anticipate and avoid potential DIs [21]. In
new APs are increasingly prescribed not only for schizophrenia principle, concomitant treatment with drugs metabolized by the
and bipolar disorder, but for adjuvant treatment of major depres- same enzyme or co-administration of a drug with another medica-
Table 1
PK parameters of AEDs. Based on [1,10,19].

74
Bioavail-ability (%) Protein binding (%) Half-life (h) Most important Other ways of Active metabolites Induction or inhibition of metabolic enzymes
way of eliminationa elimination

FIRST-GENERATION AEDs
Carbamazepine 75−85 75 5−26 CYP3A4b CYP2C8, CYP1A2, Carbamazepine Induction of CYP1A2, CYP2A6, CYP2B6,
UGT2B7, epoxide epoxide CYP2C9, CYP2C19, CYP3A4, UGT1A1, UGT2B7,
hydrolase UGT2B15, epoxide hydrolase
Ethosuximide 90−100 <10 25−70 CYP3A4b CYP2E1, renal
Phenytoin 90 90 7−80 CYP2C9b CYP2C19 (less Induction of CYP1A2, CYP2B6, CYP2C8, CYP2C9,
important: CYP2C19, CYP3A4, UGTs, epoxide hydrolase
CYP2C18, CYP3A4)
Phenobarbital >95 45−60 70−130 CYP2C9b CYP2C19, CYP2E1, Induction of CYP1A2, CYP2B6, CYP2C8,
N-glucosidation, CYP2C9, CYP2C19, CYP2E1, CYP3A4, UGT,
renal epoxide hydrolase
Primidone >90 10 5−20 CYP2C9b Renal, CYP2C19, Phenobarbital Induction of CYP1A2, CYP2B6, CYP2C8,
CYP2E1, CYP2C9, CYP2C19, CYP2E1, CYP3A4, UGT,
N-glucosidation, epoxide hydrolase
Valproic acid >90 70−95 8−16 Several UGTs ␤-oxidation, Inhibition of CYP2C9, UGT1A4, UGT2B7,
CYP2C9 (other epoxide hydrolase, N-glucosidation.
CYPs are minor) Mild auto-induction3
and mild induction of other drugsc

E. Spina et al. / Pharmacological Research 106 (2016) 72–86


SECOND-GENERATION AEDs
Clobazam ∼100 80−90 36−42 CYP3A4b CYP2C19d N- Mild induction of CYP3A4
desmethylclobazam Mild inhibition of CYP2C19 and CYP2D6
Eslicarbazepine ∼100 30 13−20 UGTs e
Renal S-licarbazepine Mild induction of CYP3A4, UGTs.
Mild inhibition of CYP2C9, CYP2C19
Felbamate 90 20−25 11−25 CYP3A4b Renal, CYP2E1, Mild induction of CYP3A4
UGTs Inhibition of CYP2C19, ␤-oxidation
Gabapentin <65 0 5−9 Renal
Lacosamide ∼100 <15 12−16 CYP2C19b Renal
Lamotrigine >95 55 25 UGT1A4 Mild induction of UGTs.
Levetiracetam 100 <10 6−8 Renal Esterase hydrolysis
in blood
Oxcarbazepine ∼100 40 7−12 Renal UGTs MHD Mild induction of CYP3A4, UGTs
(monohydroxy Mild inhibition of CYP2C19.
derivate)f
Perampanel ∼100 95 52−129 CYP3A4 UGTs Mild induction of CYP3A4
Pregabalin >90 0 5−7 Renal
Retigabine 60 80 8−10 Arylamine N-acetyl UGTs
(ezogabine) transferase-2
Rufinamide 85 30 8−12 Carboxyles-terases Mild induction of CYP3A4, UGTs
Mild inhibition of CYP2E1.
Stiripentol ≥70 99 4.5−13 CYP1A2, CYP2C19, Inhibition of CYP1A2, CYP2C19, CYP2D6,
CYP3A4, UGTsg CYP3A4.
Tiagabine ∼100 96 5−9 CYP3A4b
Topiramate ∼100 13−17 10−30 Renal CYP Mild induction of CYP3A4, ␤-oxidation,
UGT1A4.
Mild inhibition of CYP2C19.
Vigabatrin ≥50 0 4−7 Renal CYP (only 10%) Mild induction of CYP2C9.
Zonisamide ∼100 50 50−70 CYP3A4b N-acetylation
transferase, renal

AED: antiepileptic drugs; CYP: cytochrome P450; PK: pharmacokinetics; UGT: uridine diphosphate glucuronosyltransferase.
a
This refers to normal metabolism. Some subjects may not have this enzyme or have it inhibited. Other enzymes may become more important during induction.
b
Any substrate competes with others for the corresponding enzyme and inhibits it (competitive inhibition). There is data in the literature that competitive inhibition may occur at CYP1Ab, CYPbC9, CYPbC19, CYPbD6, and
CYP3A4. UGTs tend to have overlapping substrates and it is not clear that they can be subject to competitive inhibition. A good example of competitive inhibition is phenytoin in high serum concentrations (>20 mcg/ml) which
may saturate its metabolic enzymes, CYPbC9 and CYPbC19, completely or almost completely.
c
Valproic acid has been demonstrated to have mild auto-induction properties by inducing ␤-oxidation. Valproate may be an inducer of the metabolism of other medications including aripiprazole, clozapine, irinotecan and
olanzapine, but the induced enzymes are not established.
d
N-Desmethlyclobazam is mainly metabolized by CYP2C19.
e
Eslicarbazepine is a pro-drug that is activated by hepatic esterases to S-licarbazepine. S-licarbazepine is eliminated by UGTs and by the kidney (unchanged).
f
MHD is also called licarbazepine. Oxcarbazepine is a pro-drug and is activated by a cytosolic arylketone reductase that converts it to MHD, the active drug.
g
There are four metabolic pathways. No specific enzyme can be considered a major enzyme.
Table 2
PK parameters of new ADs. Based on [20].

Bioavail-ability (%) Protein binding (%) Half-life (h) Most important Other ways of Active metabolites Inhibitory effect on
way of eliminationa elimination CYP isoenzymes

SSRI
Citalopram 95 82 23−45 CYP2C19b CYP3A4, CYP2D6 CYP2D6 (weak)
Escitalopram 80 56 27 CYP2C19b CYP3A4, CYP2D6 CYP2D6 (weak)
Fluoxetine 80 95 2–4 days CYP2D6b CYP2C9, CYP2C19, Norfluoxetine CYP2D6 (potent)
CYP3A4 CYP2C9 (moderate)
CYP2C19 and
CYP3A4 (weak to
moderate)
CYP1A2 (weak)
Fluvoxamine <53 77 15−22 CYP1A2, CYP2D6 CYP1A2 and
CYP2C19 (potent)
CYP2C9 and
CYP3A4 (moderate)

E. Spina et al. / Pharmacological Research 106 (2016) 72–86


CYP2D6 (weak)
Paroxetine >64 93 10−21 CYP2D6b CYP3A4 CYP2D6 (potent)
CYP1A2, CYP2C9,
CYP2C19, CYP3A4
(weak)
Sertraline >44 98 22−36 CYP2B6 CYP2C19, CYP2C9, CYP2D6 (weak to
CYP2D6, CYP3A4 moderate)
CYP1A2, CYP2C9,
CYP2C19 and
CYP3A4 (weak)
SNRI
Desvenlafaxine 80 30 9−15 Renal UGT, CYP3A4
Duloxetine 50 >90 10−12 CYP1A2b CYP2D6 CYP2D6
(moderate)
Levomilnacipran 92 22 12 Renal UGT, CYP3A4
Milnacipran 85 13 8−10 Renal UGT, CYP3A4 CYP3A4 (weak)
Venlafaxine 92 27 5 CYP2D6b CYP3A4 Desvenlafaxine
Other newer ADs
Agomelatine <5 95 1−2 CYP1A2b CYP2C9
Bupropion 90 84 20 CYP2B6 Hydroxybupropion CYP2D6
Threohydrobupropion (moderate)
Erythrohydrobupropion
Mirtazapine 50 85 20−40 CYP2D6, CYP3A4 CYP1A2, UGTs
Reboxetine >60 97 12−16 CYP3A4b
Trazodone 65 89−95 5−9 CYP3A4b m-
chlorophenylpiperazine
(mCPP)
Vilazodone 72c 96−99 20−24 CYP3A4b CYP2C19, CYP2D6, CYP2C8 (?)
Carboxylesterase
Vortioxetine 75 98 57−66 CYP2D6b CYP3A4, CYP2C19,
CYP2C9, CYP2A6,
CYP2C8, CYP2B6

AD; antidepressant; CYP: cytochrome P450; PK: pharmacokinetics; SNRI: Serotonin and noradrenaline reuptake inhibitor; SSRI: Selective serotonin reuptake inhibitor; UGT: Uridine diphosphate glucuronosyltransferase.
a
This refers to normal metabolism. Some subjects may not have this enzyme or the enzyme is inhibited. Other enzymes may become more important during induction.
b
Any substrate competes with others for the corresponding enzyme and inhibits it (competitive inhibition). Data in the literature shows that competitive inhibition may occur at CYP1Ai, CYPiC9, CYPiC19, CYPiD6, and CYP3A4.
UGTs tend to have overlapping substrates and it is not clear that they can be subject to competitive inhibition. For example,venlafaxine can behave as a CYPiD6 competitive inhibitor.
c
Absorption is decreased when not administered with food.

75
76 E. Spina et al. / Pharmacological Research 106 (2016) 72–86

Table 3
PK parameters of new APs. Based on [19].

Bioavail-ability Protein binding Half-life (h) Most important Other ways of Active Induction or
(%) (%) ways of elimination metabolites inhibition of
eliminationa metabolic
enzymes

Amisulpride 43−48 17 12 Renal excretion Minimal


hepatic
metabolism
Aripiprazole 87 99 48−68 CYP2D6,b Dehydroaripiprazoled
CYP3A4b
Asenapine 35 95 1−2 UGT1A4,b CYP2D6 (weak)
CYP1A2b
Brexpiprazole 95 >90 91 CYP2D6,b
CYP3A4b
Cariprazine High (exact 91−97 2–4 days CYP3A4b CYP2D6 Didesmethylcaripirazine
values are not (parent drug),
reported) 2-3 weeks
(metabolite)
Clozapine 12−81 95 6−33 CYP1A2b CYP2C19, Norclozapinee
CYP3A4,
CYP2D6
Iloperidone 96 93 20−24 CYP2D6b CYP3A4 P88,f P95
Lurasidonec 9−19 99 18 CYP3A4b ID-14823
Olanzapine 60−80 93 20−70 CYP1A2,b CYP2D6, FMO
UGT1A4b
Paliperidone 28 30 24 Renal excretion Minimal
hepatic
metabolism
Quetiapine NA 83 5−8 CYP3A4b Norquetiapineg
Risperidone 68 90 3−24 CYP2D6,b CYP3A4b 9-
hydroxyrisperidone
Ziprasidone 60c 99 4−10 Aldehyde CYP3A4
oxidase

AP; antipsychotic; CYP: cytochrome P450; PK: pharmacokinetics; UGT: Uridine diphosphate glucuronosyltransferase.
a
This refers to normal metabolism. Some subjects may not have this enzyme or the enzyme is inhibited. Other enzymes may become more important during induction.
b
Any substrate competes with others for the corresponding enzyme and inhibits it (competitive inhibition). Data in the literature shows that competitive inhibition may
occur at CYP1Al, CYPlC9, CYPlC19, CYPlD6, and CYP3A4. UGTs tend to have overlapping substrates and it is not clear that they can be subject to competitive inhibition. For
example, quetiapine can behave as a CYP3A4 competitive inhibitor.
c
Absorption is decreased when not administered with food.
d
It is not definitively established in the literature how much dehydroaripiprazole contributes to antipsychotic efficacy or adverse drug reactions of aripiprazole.
e
Norclozapine does not appear to have antipsychotic efficacy but it may contribute to anticholinergic effects and hypersalivation.
f
It does not cross the blood-brain barrier. It may contribute to peripheral adverse drug reactions.
g
Some authors suggest that norquetiapine may contribute to quetiapine antidepressant properties but at this time this is only a hypothesis.

tion acting as an inhibitor or inducer involves a DI risk. A number while stiripentol is a potent inhibitor of CYP1A2, CYP2C19, CYP2D6
of drug-related (i.e., potency and concentration/dose ratio of the and CYP3A4. Some newer ADs including fluoxetine, fluvoxamine,
inhibitor/inducer, therapeutic index of the substrate, extent of paroxetine, duloxetine and bupropion, act as potent or moderate
metabolism of the substrate through the affected enzyme, presence inhibitors of various CYPs and may therefore impair the elimination
of active metabolites), patient-related (i.e., age, genetic predisposi- of AEDs metabolized via these isoforms (Table 2). Conversely, most
tion) and environmental factors (i.e., smoking) will then influence second-generation APs appear to be neither inhibitors nor inducers
the potential occurrence, magnitude, and clinical significance of a of the major drug-metabolizing enzymes and only asenapine may
metabolic DI [1]. As shown in Tables 1–3, most AEDs, new ADs and have weak CYP2D6 inhibitory properties (Table 3).
new APs are extensively metabolized via CYPs or UGTs. As many Other PK mechanisms are unlikely to mediate clinically impor-
compounds of these therapeutic classes share common metabolic tant DIs between AEDs and new psychotropic agents. To our
pathways, potential DIs may be anticipated. Furthermore, AEDs knowledge, no clinically significant DI has been described so far
may be involved in metabolically-based DIs because they act during at the absorption phase or at protein binding between com-
as inhibitors or inducers of various drug-metabolizing systems pounds of these drug classes [19,20]. DIs at the level of renal
(Table 1). Among first-generation AEDs, carbamazepine, phenytoin, excretion are rare and probably not clinically relevant as most
phenobarbital and primidone (enzyme-inducing AEDs) induce the AEDs and new psychotropic drugs are mainly eliminated by hep-
activity of CYP1A2, CYP2C9, CYP2C19 and CYP3A4, as well as UGTs atic metabolism. In theory, DIs during the excretion phase can
and epoxide hydrolase. Valproic acid is a broad-spectrum inhibitor be expected to occur only with those AEDs (i.e., gabapentin,
of various drug-metabolizing enzymes including CYP2C9 and UGTs pregabalin, vigabatrin, levetiracetam and topiramate), new ADs
and, recently, some inductive effects have been demonstrated. (i.e.,desvenlafaxine, levominalcipran and milnacipran) and second-
The newer AEDs are associated with limited enzyme-inducing generation APs (i.e., amisulpride and paliperidone) which are
potential compared with older-generation compounds. How- eliminated predominantly through the kidneys [19,20]. PK DIs
ever, clobazam, eslicarbazepine, felbamate, oxcarbazepine (at between AEDs and new ADs or APs may also involve drug
dosages ≥ 1200 mg/day), perampanel, rufinamide and topiramate transporters, in particular P-glycoprotein (P-gp), which plays an
(at dosages ≥ 400 mg/day) can induce the activity of CYP3A4 and, important role in the absorption, distribution and excretion of a
possibly, some UGTs. Some of the newer AEDs may at times act wide variety of therapeutic agents [22]. P-gp is a multidrug efflux
as enzyme inhibitors. In this respect, eslicarbazepine, felbamate, transporter highly expressed in the small intestine, brain, liver and
oxcarbazepine and topiramate are weak inhibitors of CYP2C19, kidney that acts as a natural defense mechanism against several
E. Spina et al. / Pharmacological Research 106 (2016) 72–86 77

drugs by limiting their absorption from the gut and penetration p< 0.05), respectively [37]. This effect is presumably related to non-
into the brain and promoting their elimination in the bile and urine. stereoselective induction of CYP3A4-mediated N-demethylation of
As some AEDs, ADs and APs may act as substrates, inducers or citalopram by carbamazepine.
inhibitors of P-gp [23–25], it cannot be excluded that some PK DIs In a study of healthy subjects, co-administration with carba-
between these agents, currently believed to be CYP-mediated, may mazepine, 200 mg twice a day for 32 days, was found to decrease
also be explained by modulation of multidrug-transporters [26]. by approximately 20% plasma concentrations of milnacipran, an
SNRI antidepressant whose metabolism is partly dependent on
2.2. PD DIs CYP3A4 [38]. Two PK investigations evaluated the reciprocal DI
between the new AD mirtazapine (30 mg/day) and carbamazepine
PD DIs take place directly at the site of action of a drug or indi- (400 mg/day) or phenytoin (200 mg/day) in healthy volunteers
rectly by interfering with other physiological mechanisms. They under steady-state conditions [39,40]. In the first study, the addi-
result in a modification of the pharmacological action of a drug tion of carbamazepine in 24 healthy subjects resulted in a mean
without any change in the plasma concentration and are more dif- decrease of 39% in the peak plasma concentration (Cmax ) of mirtaza-
ficult to identify and measure than pharmacokinetic DIs. These DIs pine and of 61% in the area under the plasma concentration-time
can be additive (i.e., equal to the sum of the effects of the indi- curve (AUC) [39]. Similarly, co-administration with phenytoin in 17
vidual drugs), synergistic (i.e., combined effects are greater than healthy male volunteers was associated with a mean reduction of
expected from the sum of individual effects) or antagonistic (i.e., the Cmax and the AUC of mirtazapine by 33% and 47%, respectively
combined effects are less than additive) [1]. They can be associ- [40]. Induction of CYP3A4-mediated metabolism of mirtazapine
ated with positive effects (increased efficacy and safety) or negative by carbamazepine and phenytoin may explain the observed PK
effects (decreased efficacy and/or safety). They will not be further changes.
reviewed in this article but interested readers can look at two prior A clinically relevant PK DI may occur between carbamazepine
review articles; one article described the PD DIs between AEDs and and bupropion. In a study involving 12 patients with mood dis-
ADs [20] and the other between AEDs and APs [19]. orders given a single 150-mg dose of bupropion, addition of
carbamazepine, at a mean dose of 942 mg/day, was found to
decrease the Cmax of bupropion by 87% (p < 0.001) and its AUC by
3. DIs between AEDs and new ADs 90% (p < 0.001) [41]. On the other hand, carbamazepine increased
the AUC of its pharmacologically active metabolite hydroxybupro-
AEDs and new ADs are often prescribed together in patients with pion by 50% (p < 0.05). This DI is presumably due to the inducing
different clinical conditions [20]. As the incidence of depressive effect of carbamazepine on CYP2B6, the major isoform responsible
disorders in epileptic patients is estimated to be 30%–70% in their for the metabolism of bupropion.
lifetime, ADs are frequently administered to patients with epilepsy Reboxetine, trazodone and vilazodone, which are mainly metab-
[27,28]. Some AEDs, notably valproic acid, carbamazepine and lam- olized by CYP3A4, may be particularly sensitive to induction by
otrigine, are also used as mood stabilizers and, therefore, may be enzyme-inducing AEDs. In this respect, a case report described two
given in combination with ADs for the management of bipolar dis- patients with low serum concentrations of reboxetine in relation to
order [29]. Pregabalin is also approved in Europe for the treatment the dose during carbamazepine or phenobarbital treatment, possi-
of generalized anxiety disorder and may be added to SSRI or SNRI bly mediated by induction of CYP3A4 [42].
treatment in patients with partial response [30]. Combined treat-
ment of the new AEDs, pregabalin or gabapentin, and ADs of the 3.1.2. Valproic acid
SNRI class may be a useful strategy for the management of neuro- Valproic acid has traditionally been considered a broad-
pathic pain [31,32]. spectrum inhibitor of various drug-metabolizing enzymes as
it inhibits different CYPs including CYP2C9 and, to a lesser
3.1. Effect of AEDs on the PKs of new ADs extent,CYP2C19 and CYP3A4, some UGTs, probably UGT1A4
andUGT2B7, and epoxide hydrolase [1]. Moreover, recent in vitro
3.1.1. Enzyme-inducing AEDs and in vivo evidence indicates that valproic acid may behave as a
The traditional AEDs carbamazepine, phenytoin, phenobarbital mild inducer when administered at high doses [43]. A study based
and primidone are potent inducers of several drug-metabolizing on a retrospective analysis of venlafaxine TDM data examined the
enzymes including CYPs, specifically CYP1A2, CYP2A6, CYP2B6, potential DI between valproic acid and venlafaxine, an SNRI antide-
CYP2C9, CYP2C19, CYP3A4, as well as UGTs such as UGT1A1, pressant mainly metabolized by CYP2D6 and, to a lesser extent, by
UGT2B7, and UGT2B15 [1]. Case reports and formal PK studies in CYP2C9 and CYP3A4 [44]. The mean dose-corrected serum concen-
patients or healthy subjects have repeatedly documented that con- trations of venlafaxine plus O-desmethylvenlafaxine in 41 patients
comitant treatment with enzyme-inducing AEDs may produce a receiving valproic acid co-medication did not differ significantly
potentially clinically significant decrease in drug concentrations of from those of matched controls on venlafaxine monotherapy.
most new ADs. However, the dose-corrected serum concentrations of the active
Concerning SSRIs, earlier PK investigations showed that phe- metabolite O-desmethylvenlafaxine were significantly higher (by
nobarbital, phenytoin and carbamazepine caused a decrease by 27%, p < 0.02) in patients treated with the combination of valproic
about 25% in the plasma levels of paroxetine, probably as a result of acid with venlafaxine. To explain the elevation of serum concen-
induced metabolism [33,34]. Administration of carbamazepine was trations of O-desmethylvenlafaxine, the authors speculate that the
reported to cause a marked decrease in plasma sertraline concen- inhibition of the CYP2C9-mediated N-demethylation of venlafax-
trations in two patients, resulting in lack of AD efficacy [35] and ine by valproate may lead to a stronger O-demethylation and, as
in a case-control study phenytoin or carbamazepine were asso- a consequence, to higher serum levels of O-desmethylvenlafaxine.
ciated with a sertraline C/D ratio 3 times lower [36]. In an open Differently from carbamazepine, valproic acid did not affect the
pilot study involving 6 patients with major depression stabilized metabolism of bupropion in patients with depression [41].
on citalopram (40–60 mg/day), augmentation treatment with rel-
atively low doses of carbamazepine (200–400 mg/day) for 4 weeks 3.1.3. Newer AEDs
was associated with a significant decrease in plasma concentra- Unlike first-generation agents, newer AEDs have a lower
tions of S-citalopram and R-citalopram by 27% and 31% (both, propensity to cause PK DIs [1]. Although some of the newer AEDs
78 E. Spina et al. / Pharmacological Research 106 (2016) 72–86

may act as inhibitors or inducers of specific drug-metabolizing at a dose of 200 mg/day for 17 days, did not affect the pharma-
enzymes (see previous section) and may theoretically affect AD cokinetics of carbamazepine (400 mg/day for 32 days; n = 14) and
disposition, to our knowledge, no systematic study so far has inves- phenytoin (300 mg/day for 24 days; n = 30), substrates for CYP3A4
tigated the effect of any of these AEDs on the PKs of newer ADs. and CYP2C9, respectively [58,59]. The possibility of a DI between
sertraline and valproic acid was suspected on the basis of a report
3.2. Effect of new ADs on the PKs of AEDs describing the case of a patient with bipolar depression treated
with valproic acid who developed signs of toxicity, associated with
Some new ADs act as inhibitors of various CYPs and may there- a 3-fold elevation in serum concentration of valproic acid, after the
fore impair the biotransformation of various AEDs [12,13]. addition of sertraline (100 mg/day) [60]. A clinically-significant DI
may also occur between sertraline and lamotrigine. In two epilep-
3.2.1. SSRIs tic patients treated with lamotrigine the addition of a low dose of
Fluoxetine and its metabolite norfluoxetine are potent sertraline, 25 mg/day, resulted in a two-fold elevation in plasma
inhibitors of CYP2D6 and moderate inhibitors of CYP2C9, while lamotrigine concentration associated with symptoms of toxicity,
they mildly to moderately decrease the activity of CYP2C19 and such as fatigue, sedation, confusion and decreased cognition [61].
CYP3A4 [12]. Early case reports described concomitant use of flu- Inhibition of lamotrigine glucuronidation by sertraline has been
oxetine and phenytoin as associated with significantly increased proposed to explain this DI. A subsequent study, based on a ret-
phenytoin serum levels along with signs of toxicity [45–47]. This rospective analysis of lamotrigine plasma concentrations, did not
DI can be attributed to the inhibitory effect of fluoxetine on the confirm the severity of the DI [62]. Dose-corrected concentrations
CYP2C9-mediated biotransformation of phenytoin. A number of of lamotrigine were only slightly, but not significantly, higher (by
case reports indicated that the addition of fluoxetine to an ongo- 18%) in 7 patients treated with both drugs in combination than in
ing treatment with valproic acid may increase steady-state plasma the 44 patients receiving lamotrigine without sertraline.
concentrations of valproic acid, possibly associated with toxic Citalopram and escitalopram are weak in vitro inhibitors of
effects [48–50]. The most plausible explanation for the raised val- CYP2D6 and negligible inhibitors of CYP1A2, CYP2C9, CYP2C19 and
proic acid levels is inhibition of CYP2C9 by fluoxetine. There is CYP3A4 [12,13]. Due to their minimal effect on drug-metabolizing
conflicting evidence for a PK DI between fluoxetine and carba- enzymes, citalopram and escitalopram are not expected to cause
mazepine. Although case reports have documented that fluoxetine clinically relevant DIs with AEDs. Consistent with this, in an
increased carbamazepine levels [51,52], no changes in steady-state open-label investigation in 12 healthy volunteers, the addition of
concentrations of carbamazepine were observed in eight epileptic citalopram, 40 mg/day for 14 days, to carbamazepine, administered
patients stabilized on carbamazepine (800–1600 mg/day) after a at the dose of 400 mg/day for 35 days, did not alter the steady-state
3-week co-administration with fluoxetine, 20 mg/day [53]. It can PK parameters of carbamazepine and its active epoxide metabolite
be speculated that inhibition of CYP3A4-mediated carbamazepine [63].
metabolism might occur only at higher fluoxetine doses or require
more than 3 weeks to manifest, due to the long half-life of norfluox-
etine. Retrospective analysis of a large lamotrigine TDM database 3.2.2. SNRIs
revealed that dose-normalized serum concentrations of lamotrig- Among SNRIs, venlafaxine, desvenlafaxine, milnacipran and
ine were 39% lower inpatients co-prescribed fluoxetine (n = 15) as levomilnacipran are negligible inhibitors of CYP isoforms (or min-
compared to patients receiving lamotrigine monotherapy (n = 614) imally affect the activity of CYPs), while duloxetine is a moderate
[54]. As fluoxetine is not known as an enzyme inducer, it is difficult inhibitor of CYP2D6 [13]. To date, there have been no reports that
to explain the observed effect on lamotrigine. these ADs affect the PKs of AEDs.
Paroxetine is a potent inhibitor of CYP2D6, while it only min-
imally affects other CYPs [12]. As CYP2D6 plays a minor role in
the biotransformation of AEDs, paroxetine is not expected to cause 3.2.3. Other newer antidepressants
clinically-relevant DIs with these agents. Consistent with this, a Mirtazapine has minimal inhibitory effects on the various CYP
single-blind, placebo-controlled, cross-over study of 20 patients isoforms in vitro [13]. In agreement with this, two PK studies
with epilepsy, stabilized on phenytoin, carbamazepine or valproic in healthy subjects showed that mirtazapine (30 mg/day) had
acid, showed that addition of paroxetine, 10–30 mg/day for 16 days, no effect on the disposition of both carbamazepine and pheny-
did not affect significantly plasma concentrations of these AEDs toin [39,40]. Trazodone, a CYP3A4 substrate, has no significant
[34]. inhibitory effect on the activity of CYP isoforms. The addition of tra-
Fluvoxamine is a potent inhibitor of CYP1A2 and CYP2C19, zodone in a patient receiving a chronic treatment of carbamazepine
a moderate inhibitor of CYP2C9 and CYP3A4, and only a weak was associated with symptoms of carbamazepine toxicity, accom-
inhibitor of CYP2D6 [12]. Being an inhibitor of different CYP iso- panied by an increase in its serum concentration [64]. After the
forms, fluvoxamine may theoretically impair the elimination of discontinuation of trazodone, toxic symptoms lessened and carba-
various AEDs. The possibility of a clinically relevant DI between mazepine concentrations progressively decreased, suggesting the
fluvoxamine and carbamazepine was suggested by case reports possibility of a PKDI between the two drugs. Competitive inhibition
describing increased carbamazepine levels with associated symp- of CYP3A4 may provide a possible explanation for this case.
toms of toxicity following the addition of fluvoxamine [55,56]. Anecdotal evidence indicates that bupropion may inhibit the
However, a study of seven epileptic patients on a chronic treatment metabolism of phenytoin and valproate [65]. On the other hand, in
with carbamazepine (800–1600 mg/day) found no modifications in a randomized, open-label, two-way crossover study in 12 healthy
carbamazepine levels after three weeks of co-administration with subjects, PK parameters of a single oral dose of 100 mg lamotrig-
fluvoxamine, 100 mg/day [53]. A case report documented a 3-fold ine were not affected by steady-state bupropion, 150 mg twice/day
increase in serum concentrations of phenytoin after administra- [66].
tion of fluvoxamine [57]. This PK change was explained as a result Other newer ADs, such as agomelatine, vortioxetine, or vila-
of inhibition of both CYP2C9 and CYP2C19 by fluvoxamine. zodone, are only weak inhibitors or are not inhibitors of CYP
Sertraline is a mild to moderate in vitro inhibitor of CYP2D6 isoforms at usual therapeutic concentrations and are not expected
and a weak inhibitor of the other CYP isoenzymes [12]. Two for- to affect thedisposition of concomitantly administered AEDs.
mal PK studies in healthy volunteers documented that sertraline,
E. Spina et al. / Pharmacological Research 106 (2016) 72–86 79

4. DIs between AEDs and new APs [81,82]. A subsequent study showed that risperidone active moi-
ety (sum of plasma concentrations of risperidone and its active
AEDs and new APs may be used in combination for a number of 9-hydroxyrisperidone metabolite) was approximately 1.4 times
reasons [17,19]: (a) there is a high prevalence of psychotic disorders lower in 11 carbamazepine patients versus 23 patients on risperi-
in patients with epilepsy; (b) some AEDs, particularly those with done alone [83]. In 5 patients assessed on and off carbamazepine
mood-stabilizing properties (i.e., carbamazepine, valproic acid and co-medication, risperidone active moiety was 1.5 times lower on
lamotrigine), are increasingly used in the management of bipolar carbamazepine [83]; in 11 other patients it was 1.8 times lower
disorder; (c) autism spectrum disorders are associated with very [84].
high risk of epilepsy and some second-generation APs are approved
for the treatment of irritability in children with these disorders; (d) 4.1.1.5. Paliperidone. The new AP paliperidone is predominantly
AEDs may be used to treat dosage-related seizures associated with excreted unchanged through the kidney and undergoes limited
some new APs such as clozapine; (e) AEDs may be used to treat non- hepatic metabolism. Based on this, CYP inhibitors or inducers
epileptic conditions (e.g., neuropathic pain and migraine) which are not expected to have a significant influence on paliperi-
may occur in psychotic patients. done pharmacokinetics. In a study of six schizophrenic patients
initially treated with a 6–12 mg/d dose of paliperidone alone,
4.1. Effect of AEDs on the PKs of new APs coadministration with carbamazepine, 600 mg/day for 2–4 weeks,
was associated with an average reduction in plasma paliperi-
4.1.1. Carbamazepine done concentrations by 66% [85]. This effect may be attributed
Being a powerful inducer of several drug-metabolizing enzymes, to carbamazepine induction of drug-metabolizing enzymes, i.e.,
carbamazepine has been reported to cause a clinically significant CYP3A4, and/or drug transporters, i.e., renal P-glycoprotein. In
decrease in plasma concentrations of many second-generation APs. a PK investigation in healthy subjects, sub-therapeutic doses
of carbamazepine,400 mg/day, for 3 weeks decreased the mean
4.1.1.1. Clozapine. In a study based on a clozapine TDM database, steady-state serum peak concentration (Cmax ) and the AUC of
patients co-treated with carbamazepine had a mean 50% lower paliperidone by approximately 37%, due to a 35% increase in the
concentration-to-dose (C/D) ratio of clozapine than the monother- renal clearance of paliperidone [86].
apy group [67]. Moreover, in a study of 12 patients stabilized on
clozapine therapy, serum concentrations of clozapine were 47%
4.1.1.6. Ziprasidone. In a study of 19 healthy volunteers receiving
lower in patients also treated with carbamazepine than in those
40 mg/day of ziprasidone, carbamazepine (400 mg/day) lowered
receiving oxcarbazepine [68]. The inducing effect of carbamazepine
ziprasidone mean Cmax by 27% and mean AUC0-12h by 36% [87].
on CYP1A2- and CYP3A4-mediated clozapine metabolism probably
These changes are probably due to an inducing effect of carba-
explains these results.
mazepine on CYP3A4-mediated metabolism of ziprasidone.

4.1.1.2. Olanzapine. The pharmacokinetics of a single 10-mg olan-


4.1.1.7. Aripiprazole. Co-administration of carbamazepine for 4–6
zapine dose were investigated in 11 healthy volunteers before and
weeks in 9 patients with schizophrenia or schizoaffective disorder
after administration of carbamazepine 400 mg/day for 18 days [69].
given aripiprazole monotherapy (30 mg/day) caused a significant
Carbamazepine caused a 46% increase in olanzapine clearance and a
decrease in the AUC of aripiprazole and dehydroaripiprazole serum
34% decrease in its AUC. A number of subsequent TDM studies doc-
concentrations by 71% and 69%(both, p< 0.001), respectively, [88].
umented that the median C/D ratio of olanzapine was 36–71% lower
Similarly, in a study of 18 schizophrenia patients treated with arip-
in carbamazepine patients than in those on olanzapine monother-
iprazole (12 or 24 mg/day), a 1-week addition of carbamazepine,
apy [70–74]. In their study, Linnet and Olesen [71] measured
400 mg/day, significantly decreased plasma concentrations of both
both free and glucuronidated olanzapine and found that carba-
aripiprazole and dehydroaripiprazole by 64% and 68% (p< 0.001),
mazepine lowered free olanzapine concentration by increasing
respectively [89]. Consistent with these findings, a routine TDM
glucuronidation metabolism. All of these studies clearly indicated
study indicated that carbamazepine lowered dose-adjusted con-
that carbamazepine increases olanzapine metabolism by CYP1A2-
centrations of aripiprazole by 88% [90].
and UGT-induction.

4.1.1.3. Quetiapine. In a PK study of 18 psychiatric patients, car- 4.1.1.8. Other new APs. Carbamazepine is expected to induce the
bamazepine (600 mg/day) decreased quetiapine Cmax by 80% and metabolism of other recently commercialized APs such as lurasi-
AUC by 87% and causeda 7.5-fold increase in its oral clearance [75]. done and cariprazine, mainly metabolized by CYP3A4, iloperidone
Retrospective evaluation of quetiapine TDM databases provided and brexpiprazole, partially metabolized by CYP3A4, and asenap-
similar results [76–79]. In particular, in a large TDM study, co- ine, substrate for CYP1A2 and UGT1A4. To date, no study has
administration with carbamazepine (n = 39) was associated with investigated the effect of carbamazepine on the PKs of these new
a significant decrease by 86% (p < 0.001) in quetiapine C/D ratio as medications.
compared to quetiapine monotherapy (n = 1123) [77]. The clinical
implications of this DI were demonstrated in three patients tak- 4.1.2. Phenobarbital/phenytoin
ing carbamazepine 400–800 mg/day in which serum quetiapine Like carbamazepine, phenobarbital and phenytoin are consid-
concentrations could not be detected (<25 ng/mL) despite taking ered powerful inducers of CYPs, (including CYP1A2, CYP2C, and
700 mg/day [80]. These massive PK changes are explained by the CYP3A4) as well as UGTs.
potent inducing effect of carbamazepine on CYP3A4-mediated que- Phenobarbital and phenytoin may cause a clinically relevant
tiapine metabolism. Appropriate dosage adjustment of quetiapine induction of clozapine metabolism. Addition of phenytoin to a
is obviously needed if the two drugs are given concurrently. stable clozapine treatment resulted in a psychotic exacerbation
associated with decreased clozapine levels by 65–85% [91]. A
4.1.1.4. Risperidone. Two case reports suggested the possibility remarkable elevation of clozapine concentrations was described
of a clinically relevant PK interaction between carbamazepine in a patient 2 weeks after phenobarbital discontinuation [92].
and risperidone, presumably due to an inducing effect of carba- In a study of patients with schizophrenia, serum clozapine con-
mazepine on CYP3A4-partly dependent risperidone metabolism centrations were significantly lower in 7 phenobarbital-treated
80 E. Spina et al. / Pharmacological Research 106 (2016) 72–86

patients as compared to 15 patients on clozapine monotherapy lower (−32%, p < 0.001) in patients co-medicated with valproic acid
(232 ± 104 ng/ml versus 356 ± 13 ng/ml) [93]. (n = 92), compared with controls taking olanzapine alone (n = 205).
A clinically significant PK DI has been reported to occur These changes are likely to be explained by valproic acid induction
between phenytoin and quetiapine. In a study of 10 patients on olanzapine metabolism by CYPs or UGTs with the possibility of
treated with quetiapine, 750 mg/day, co-administration of pheny- added competitive inhibition. The mechanisms and clinical rele-
toin, 300 mg/day, decreased plasma quetiapine concentrations by vance of this DI need to be better investigated.
approximately 80%, reflecting a potent inducing effect of phenytoin
on CYP3A4-mediated quetiapine biotransformation [94]. 4.1.3.3. Quetiapine. Studies on the effect of valproic acid on que-
tiapine PKs provided inconsistent results. A TDM study found that
4.1.3. Valproic acid quetiapine C/D ratios were 77% higher (p = 0.016) in patients co-
As valproic acid is increasingly used in the treatment of bipolar medicated with valproic acid as compared to those not receiving
disorder, in recent years several studies investigated its effect on valproic acid [107]. Based on this finding, the authors suggested a
the PKs of different second-generation APs. CYP3A4-mediated inhibition of quetiapine metabolism by valproic
acid. However, co-administration with valproic acid was associated
4.1.3.1. Clozapine. Four studies reported a slight increase in cloza- with no relevant changes in quetiapine concentrations accord-
pine concentrations following co-administration of valproic acid, ing to two other routine TDM studies [77,78] and a formal PK
suggesting that valproic acid may inhibit clozapine conversion to study in 34 psychotic patients taking 300 mg/day of quetiapine and
norclozapine via CYP1A2 or CYP3A4 [95–98]. Conversely, in a case 1000 mg/day of divalproex sodium [108].
series of four patients, clozapine concentrations were reported to
decrease by a mean 41% when valproic acid was administered 4.1.3.4. Risperidone. No major changes in risperidone moiety
[99]. By using a statistical mixed model that estimated the effect were found in a PK study comparing 23 patients on risperi-
sizes of co-medications on clozapine levels after correcting for done monotherapy vs 10 on valproic acid co-medication
confounding variables, Diaz et al. [100] found that valproic acid (1200–1500 mg/day) [83], and in another PK investigation of 22
appeared to act as a clozapine inhibitor in non-smokers and as bipolar patients taking valproic acid (1000 mg/day) in which half
an inducer in smokers. These findings may account for the con- were randomized to placebo or risperidone up to 4 mg/day [109].
troversial results of the earlier observations. A replication with This is consistent with the absence of an inhibitory effect of val-
the same statistical model in another sample, verify mild induc- proic acid on CYP2D6 or CYP3A4, the major isoforms involved in
tive effects of valproic acid on clozapine [101]. A case report in risperidone metabolism.
a smoker suggests that on rare occasions valproic acid can have
major inductive effects on clozapine, similar to those of potent AED
4.1.3.5. Aripiprazole. In a PKstudy of 10 patients with schizophre-
inducers that may be dose-related [102]. Similarly, case reports and
nia, valproic acid co-administration resulted in decreases in the
case-control studies suggest that the inhibitory effects of valproic
Cmax and AUC of aripiprazole by 26% and 24%, respectively, with
acid on clozapine metabolism may have relevance by accelerating
minimal effects on PK parameters of the active metabolite dehy-
already rapid clozapine titrations and contributing to clozapine-
droaripiprazole [110]. Similarly, in a TDM study, aripiprazole C/D
induced myocarditis by a mechanism analogous to the contribution
ratio was 24% lower in 7 patients co-medicated with valproic acid
of valproic acid to lamotrigine-induced Stevens-Johnson syndrome
than in 23 on aripiprazole monotherapy [90]. As aripiprazole is a
[103].
CYP3A4 and P-gp substrate, these effects may be explained by a
mild induction of valproic acid on CYP3A4 and/or P-gp.
4.1.3.2. Olanzapine. A PK interaction may theoretically occur
between valproic acid and olanzapine, as both compounds are
metabolized, at least partially, by UGT1A4. Initial TDM studies 4.1.3.6. Asenapine. The metabolism of asenapine is predominantly
found no significant effect of valproic acid on plasma olanzapine mediated by UGT1A4 and CYP1A2. In an open-label, randomized, 2-
concentrations [73,104]. However, in four patients stabilized on way crossover study in 24 healthy male volunteers receiving 5 mg
olanzapine, valproic acid (1000–2700 mg/day) decreased olanza- of asenapine alone or under steady-state valproate (1000 mg/day
pine levels by 50% with a psychiatric deterioration in three of them for 9 days), valproic acid substantially reduced the formation of
[105].A subsequent study investigated the effect of valproic acid on the N-glucuronide (AUC reduced 7.4-fold) and moderately reduced
the steady-state plasma concentrations of olanzapine in 18 patients that of N-desmethyl-asenapine (AUC reduced 30%) but did not
with bipolar or schizoaffective disorder [106]. After 4 weeks of affect asenapine AUC [111]. The authors concluded that low-dose
valproic acid co-administration (600–2000 mg/day), mean serum valproic acid, although almost completely inhibiting asenapine glu-
olanzapine concentrations were decreased by 18%, but no patient curonidation, did not affect the PKs of asenapine itself.
showed a worsening of psychopathological condition. The use of
a statistical model helped to clarify the clinical relevance of the 4.1.4. Lamotrigine
results of this prospective study in spite of their complexity [43]. Lamotrigine is a weak UGT inducer, while it does not appear
The changes on olanzapine concentration varied with time (from to be a CYP inhibitor or inducer [1]. In view of the frequent com-
2 to 4 weeks) and with valproic acid concentrations, and could bination of lamotrigine with second-generation APs, a number of
be best interpreted as valproic acid being both an inducer and a studies have examined the possibility of a PKDI between these
competitive inhibitor of olanzapine metabolism. The model could compounds.
be used to predict the effects of any olanzapine dose at 2 and
4 weeks. If we focus on 10 mg/day of olanzapine at 4 weeks as 4.1.4.1. Clozapine. A case report described the occurrence of symp-
an example: 1) in nonsmokers, valproate was an net inhibitor at toms of clozapine toxicity associated with a 3-fold increase in
concentrations <42 ␮g/mL since the competitive inhibition over- serum clozapine concentrations in a patient after the addition of
come the inductive effects; and 2) in smokers valproate was an lamotrigine [112]. However two subsequent studies, the first in 34
net inhibitor only at concentrations <13 ␮g/mL, beyond this low hospitalized treatment-resistant patients [113] and the second in
valproate concentrations the inductive effects were greater than 11 patients stabilized on clozapine [114], found no serum cloza-
the effects of the competitive inhibition. In a more recent TDM pine concentration changes during lamotrigine treatment up to
study [74], serum concentrations of olanzapine were significantly 200 mg/day.
E. Spina et al. / Pharmacological Research 106 (2016) 72–86 81

Table 4
Provisional new AD dose correction factors after adding AEDs and AED dose correction factors after adding new ADs based on the limited available information.

AEDs ADs Outcome Action to take

Powerful inducers ↓ AD level 2–4 weeks In the absence of TDM, use correction
carbamazepine, after adding inducer factor:a
phenobarbital Bupropion ↑ AD level 2–4 weeks Avoid: correction factor is 10
phenytoin, primidone Sertraline after D/C inducer Correction factor: 3 (up to 5 in case
reports)
Mirtazapine Correction factor: 2–3
Citalopram, milnacipran, paroxetine Correction factor is only 1.3. If this
correction factor is correct, it may not
be clinically relevant.
Trazadone, reboxetine, vilazodone Be careful; induction of these ADs that
are dependent on CYP3A4 for their
metabolism may require high
correction factors: ≥5.
Agomelatine, duloxetine Not studied but likely to be clinically
relevant according to CYP involved in
AD metabolism
Desvenlafaxine, escitalopram, Not studied but unlikely to be relevant
fluoxetine, fluvoxamine, according to CYP involved in AD
levomilnacipran, metabolism
venlafaxine,vortioxetine
High dose mild inducers: Possibly relevant for: bupropion, Mild ↓ ADlevel weeks after adding Monitor for ↓ efficacy after adding mild
oxcarbazepine (≥1200 mg/day) or mirtazapine, reboxetine, sertraline, mild inducer inducer.
topiramate (≥400 mg/day). And trazadone, vilazodone Mild ↑ AD level weeks after D/C mild Monitor for ADRs after discontinuing
probably high doses of clobazam, inducer mild inducer.
eslicarbazepine, felbamate or
rufinamide.
Phenytoin, valproic acid Inhibitor: Fluvoxamine ↑ AED level after adding fluvoxamine It is safer to select another AD.
Not studied but likely to be relevant: ↓ AED level after D/C fluvoxamine Use AED TDM if you decide to add
clobazam, lacosamide, phenobarbital, fluvoxamine.
primidone
Not studied; but possible relevant:
carbamazepine, felbamate, tiagabine,
zonisamide
Phenytoin, valproic acid Inhibitor: Fluoxetine ↑ AED level 2–3 months after adding It is safer to select another AD.
Not studied at steady state; but likely fluoxetine in average patient (some Use AED TDM if you decide to add
relevant: clobazam, lacosamide, may take longer) fluoxetine.
phenobarbital, primidone ↓ AED level 2–3 months after adding
Not studied at steady state; but fluoxetine in average patient (some
possible relevant: carbamazepine, may take longer)
felbamate, tiagabine, zonisamide

AD: antidepressant; AED: antiepileptic drug; D/C: discontinuing; DI: drug–drug interaction; TDM: therapeutic drug monitoring.
a
Reviewed PK DI studies were used to estimate correction factors, which are >1 in inducers and <1 in inhibitors. A correction factor of 2 indicates that you need to double
the dose for the same effect. A correction factor of 0.5 indicates that you need to halve the dose for the same effect. The further the correction factor is from r, the more
relevant the DI may be. The therapeutic window or index also determines the clinical relevance of correction factors.

4.1.4.2. Olanzapine. Although UGT1A4 metabolizes both lamot- ine enhances the metabolism of quetiapine, probably via an action
rigine and olanzapine, two studies of healthy volunteers using on UGT1A4 glucuronidation.
low lamotrigine doses (50 and 200 mg/day) found no effects on
olanzapine PKs [115,116]. A subsequent study of 14 patients 4.1.4.4. Risperidone. A case report described the occurrence of
taking olanzapine (10–20 mg/day) showed that lamotrigine at risperidone adverse effects associated with a 5–6-fold elevation
a dosage of 100 mg/day did not modify the PKs of olanzapine, in risperidone levels after adding lamotrigine [118]. However, a
while mean plasma olanzapine concentrations increased by 16% prospective study of 10 psychotic patients stabilized on risperi-
at a lamotrigine dosage of 200 mg/day [114]. By using a mixed done (3–6 mg/day), demonstrated that lamotrigine, titrated up to
model, Botts et al. confirmed that lamotrigine behaved as an a final dosage of 200 mg/day over 8 weeks, did not affect serum
olanzapine inhibitor in smokers, increasing the C/D ratio by 35%, risperidone active moiety [114].
whereas lamotrigine produced a mild non-significant decrease in
olanzapine C/D ratio in nonsmokers [73]. However, in a retro-
4.1.4.5. Aripiprazole. Analysis from an aripiprazole TDM database
spective analysis of olanzapine TDM data, serum concentration
found no significant differences in serum concentrations of arip-
of olanzapine did not differ between patients treated with lam-
iprazole and dehydroaripiprazole in patients who were prescribed
otrigine (n = 110) and the control group on olanzapine alone
lamotrigine concomitantly as compared to those on aripiprazole
(n = 205) [74].
alone [119].

4.1.4.3. Quetiapine. A large TDM study reported a slight (17%) but 4.1.5. Oxcarbazepine
significant decrease in quetiapine C/D ratio in 147 lamotrigine Oxcarbazepine, a keto-analog of carbamazepine, is a weak
patients when 1123 were on quetiapine monotherapy [77].Analysis inducer of CYP3A4 and glucuronidation enzymes [1]. Concomitant
of data from a subsequent quetiapine TDM database found that treatment with oxcarbazepine (900–1200 mg/day) for 5 weeks
mean plasma quetiapine C/D ratios were 57% lower in 22 patients in psychotic patients stabilized on risperidone (2–6 mg/day;
who were on concomitant lamotrigine, compared with matched n = 12) or olanzapine (5–20 mg/day; n = 13) was associated with
controls who were not [117]. The authors concluded that lamotrig- minimal and not significant changes in serum concentration
82 E. Spina et al. / Pharmacological Research 106 (2016) 72–86

Table 5
Provisional new AP dose correction factors during PK DIs with AEDs with inhibitory and inducing properties based on the limited available information.

APs AEDs (inducers or inhibitors) Outcome Actions to takea

CYP1A2 APs Potent inducers: ↓ AP level 2–4 weeks after adding 1) Correction dose factor:
Clozapine carbamazepine, phenytoin or phenobarbital. inducer Clozapine: 1.5–2. Olanzapine:
Olanzapine ↑ AP level 2–4 weeks after D/C 2–3
inducer 2) TDM is strongly
recommended.
3) In some countries
carbamazepine is not
recommended since it is also
associated with
agranulocytosis.
High dose mild inducers: oxcarbazepine Mild ↓ AP level weeks after adding 1) Not well studied.
(≥1200 mg/day) or topiramate (≥400 mg/day) mild inducer 2) TDM is strongly
And probably highdoses of clobazam, Mild ↑ AP level weeks after D/C recommended.
eslicarbazepine, felbamate or rufinamide. mild inducer
Valproic acid: possible mild inhibitor and/or Mild ↑ or ↓ AP level after adding 1) Not well studied.
mild inducer. valproic acid 2)TDM is strongly
Mild ↓or ↑ AP level after D/C recommended.
valproic acid
CYP3A4 APs Potent inducers: carbamazepine, phenytoin or Severe ↓ AP level 2–4 weeks after Do not use (Correction factor
Cariprazine phenobarbital. adding inducer ≥5).
Lurasidone High dose mild inducers: oxcarbazepine Unknown ↓ AP level weeks after Do not use unless access to
Quetiapine (≥1200 mg/day) or topiramate (≥400 mg/day). adding mild inducer TDM is available.
And probably high doses of clobazam, Unknown ↑ AP level weeks after
eslicarbazepine, felbamate or rufinamide. D/C mild inducer
CYP2D6/CYP3A Potent inducers: carbamazepine, phenytoin or ↓ AP level 2–4 weeks after adding 1) Correction dose factor: 2.0
APs phenobarbital. inducer 2) Use TDM if available.
Aripiprazole ↑ AP level 2–4 weeks after D/C
Brexpiprazole inducer
Iloperiodone High dose mild inducers: oxcarbazepine Mild ↓ AP level weeks after adding 1) Not well studied.
Risperidone (≥1200 mg/day) or topiramate (≥400 mg/day). mild inducer 2) Use TDM if available.
And probably high doses of clobazam, Mild ↑ AP level weeks after D/C
eslicarbazepine, felbamate or rufinamide. mild inducer
Only Aripiprazole Valproic acid: possible inducer Mild ↓ aripiprazole after adding 1) Correction dose factor: 1.25.
valproic acid 2) Consider TDM.
Mild ↑ aripiprazolelevel after D/C
valproic acid
Aldehyde oxidase Potent inducers: carbamazepine, phenytoin or Mild ↓ AP level 2–4 weeks after 1)No need for dose change
(CYP3A4) phenobarbital. adding inducer (very small correction factor
Ziprasidone Mild ↑ AP level 2–4 weeks after 1.33)
D/C inducer 2)TDM can help to verify that.
UGT1A4 & CYP1A2 Potent inducers: carbamazepine, phenytoin or Unknown 1) Not well studied.
Asenapine phenobarbital. 2)TDM is strongly
recommended.
High dose mild inducers: oxcarbazepine Unknown 1) Not well studied.
(≥1200 mg/day) or topiramate (≥400 mg/day). 2) Consider TDM.
And possibly high doses of clobazam,
eslicarbazepine, felbamate or rufinamide.
Valproic acid: possible inhibitor. Unknown 1) Not well studied. 2)
Consider TDM.
Renally excreted Potent inducers: carbamazepine, phenytoin or Not relevant 1)No need for dose change.
Amisulpride phenobarbital. 2)TDM can help to verify that.
Renally excreted & P-gp or Potent inducers: carbamazepine, phenytoin or ↓ AP level 2–4 weeks after adding 1) Correction dose factor: 3.
CYP3A4b Paliperidone phenobarbital. inducer 2)TDM is strongly
↑ AP level 2–4 weeks after D/C recommended.
inducer
High dose mild inducers: oxcarbazepine Unknown ↓ AP level weeks after 1) Not well studied.
(≥1200 mg/day) or topiramate (≥400 mg/day). adding mild inducer 2)TDM is strongly
And possibly high-doses of clobazam, Unknown ↑ AP level weeks after recommended.
eslicarbazepine, felbamate or rufinamide. D/C mild inducer
Valproic acid: possible inhibitor ↑ AP level after adding valproic acid 1) Correction factor 0.50.
↓ AP level after D/C valproic acid 2) Consider TDM.

ADRs: Adverse dug reaction; AED: antiepileptic drug; AP: antipsychotic; CYP: cytochrome P450; D/C: discontinuing; DI: drug–drug interaction; TDM: therapeutic drug
monitoring.
a
Reviewed PK DI studies were used to estimate correction factors, which are >1 in inducers and <1 in inhibitors. A correction factor of 2 indicates that you need to double
the dose for the same effect. A correction factor of 0.5 indicates that you need to halve the dose for the same effect. The further the correction factor is from s, the more
relevant the DI may be. The therapeutic window or index also determines the clinical relevance of correction factors.
b
During induction there is a major increase in the percentage of paliperidone metabolized. It is currently unclear whether this is due to P-gp, CYP3A4, or another CYP.

of these APs [120]. A recent case report suggested that long- (≥1200 mg/day) prescribed for months are needed to defini-
term treatment with a high dose oxcarbazepine (900 mg/day tively establish the effects of oxcarbazepine in second-genera-
in an 8-year-old child) can reduce quetiapine C/D ratio by tion APs.
70% [121]. Prospective studies using high oxcarbazepine doses
E. Spina et al. / Pharmacological Research 106 (2016) 72–86 83

4.1.6. Topiramate entation to clinicians with no TDM access and may also encourage
Topiramate is a weak inhibitor of CYP2C19 and a weak clinicians to further study TDM.
inducer of CYP3A4 [1].An open-label study of 12 healthy sub- Tables 4 and 5 provide correction factors calculated using the
jects investigated the effect of topiramate (100 mg twice daily) available studies, assuming that the average patient represents the
on the PKsof single-dose risperidone (2 mg) [122]. Topiramate population well. However, there is extensive PK variability among
increased mean risperidone clearancevalues by 51% and decreased patients, and TDM measurements should be advised for optimal
mean plasma risperidone AUC values by 23%. Mean plasma 9- titration of the dosage in each individual. There will be a need
hydroxyrisperidone(the pharmacologically active metabolite of to periodically update these dose correction factors provided by
risperidone) AUC values were concurrently decreased by 8%. On Tables 4 and 5 as new knowledge becomes available. These limited
the other hand, in a study involving 38 outpatients with psy- correction factors are useful because: (i) many clinicians have no
chotic disorders, steady-state plasma concentrations of clozapine access to TDM; (ii) there is limited clinical data on TDM for new
(10 patients), olanzapine (12 patients), risperidone (9 patients), or compounds; and (iii) having the correction factor helps clinicians
quetiapine (7 patients) were not significantly affected after 8 weeks make decisions to select safer drugs, with less risk of PK DDIs.
of concomitant treatment with therapeutic doses of topiramate There is relatively well understanding of the pharmacological
(up to 200 mg/day) [123]. Prospective studies using high topira- mechanisms and clinical relevance associated with the inductive
mate doses (≥400 mg/day) prescribed for months are needed to properties of carbamazepine, phenytoin, phenobarbital and prim-
definitively establish the effects of topiramate in new APs. idone on new ADs and new APs, and of the inhibitory properties
of valproic acid on some new ADs. On the other hand, PK DI stud-
4.1.7. Other newer AEDs ies combining new AEDs, particularly those with mild inductive
To date, no clinically significant PK DI involving other new AEDs properties, with new ADs and new APs are needed. It appears safer
and second-generation APs have been reported. to overcaution clinicians about the potential risks of losing efficacy
when adding inducers or of increased ADRs when adding inhibitors,
as well as the risks of ADRs when discontinuing inducers or losing
4.2. Effect of new APs on the PKs of AEDs efficacy when discontinuing inhibitors. Clinicians cannot afford to
wait for definitive studies, which are not likely to happen once the
Few studies have investigated the effect of new APs on the PKs new drugs are already marketed.
of AEDs. Using the limited available information, the authors believe
Two weeks after the addition of risperidone (1 mg/day) the that in high doses oxcarbazepine (≥1200 mg/day) and topiramate
mean steady-state carbamazepine concentrations mildly increased (≥400 mg/day) may be clinically relevant CYP3A4 inducers and
by 19% in 8 epileptic patients [124], probably due to competitive possibly inducers of some UGTs. It is possible that clobazam, esli-
inhibition of CYP3A4. Adding quetiapine to carbamazepine led to a carbazepine, and rufinamide may also be dose-dependent CYP3A4
pharmacokinetic drug interaction and adverse effects in 2 patients inducers. Two other mild inducers, felbamate and vigabatrin, are
[125], probably due to the competitive inhibition of CYP3A4. In a rarely prescribed AEDs. More problematic is the case of valproic
study involving 28 healthy volunteers, the addition of aripiprazole acid, a drug traditionally considered a potential inhibitor, which
30 mg/day to valproic acid (500 mg every 12 h) had no effect on may also be a dose-dependent mild inducer of some new APs.
valproic acid PKs [126]. According to a TDM study, PK parameters The metabolic enzymes and/or transporters influenced by valproic
of lamotrigine were not significantly modifiedby clozapine, risperi- acid induction are not known. These metabolic enzymes and/or
done orolanzapine co-prescription [127]. Likewise, in a study of 18 transporters may also be subject to competitive inhibition by val-
patients, aripiprazole (30 mg/day at steady state), did not affect sig- proic acid. Regarding valproic acid, clinicians need to be aware that
nificantly lamotrigine PKs (100–400 mg/day at steady state) [128]. it is: (1) definitely an inhibitor and/or inducer of clozapine and
In summary, new APs are not considered clinically relevant olanzapine with potential for clinically relevant DIs, (2) possibly
except asenapine, which is a weak inhibitor of CYP2D6, a metabolic an inhibitor of paliperidone, and (3) a possible weak inducer of
enzyme not relevant for AEDs. In most circumstances new APs are aripiprazole. Studies by independent investigators not associated
not expected to influence serum AED concentrations in a clinically with the pharmaceutical company are needed for determining the
relevant way. On the other hand, clinicians need to know that in effects of potent inducers and valproic acid on asenapine. More DI
rare circumstances, new APs may be clinically relevant inhibitors of quetiapine studies are needed to establish the potential of lamot-
AEDs, particularly in situations of polypharmacotherapy, possibly rigine to induce quetiapine and of valproic acid to inhibit quetiapine
due to the saturation of several CYPs by competitive inhibition. metabolism. These possible effects are not described in Table 4 until
more definitive quetiapine DI studies are published. Quetiapine
5. Conclusion TDM studies are complicated by the short half-life of this com-
pound with extreme variability within a day and between troughs
It is not easy to conduct PK DI studies using clinically relevant and peaks, making it difficult to eliminate variability due to time
doses of inducers or inhibitors. To get approval for the marketing differences in collectingsamples. Stiripentol is a clinically relevant
of their drugs, pharmaceutical companies tend to invest in simple inhibitor of several CYPs with potential to decrease the metabolism
PK DI studies in volunteers, frequently using single dosing, but this of several new APs and new ADs, but it is not likely to be combined
type of PK study rarely reflects clinical practice [129]. The DI mate- with them and there are no DI studies.
rial in prescribing information (or package inserts) is usually not Fluoxetine and fluvoxamine are relevant inhibitors of phenytoin
designed to provide easy helps for clinicians to correct PK DIs with and valproic acid, but the literature does not provide informa-
dose changes [129]. tion on calculating dose correction factors. Therefore, fluoxetine
To fill the need to orient clinicians, we have updated prior and fluvoxamine should not be added or discontinued from
tables [19,20] that integrate the information on in vitro metabolism patients taking phenytoin or valproic acid without close monitor-
studies, TDM studies, case report/series and prospective studies to ing of the antiepileptic TDM. Other not-well-studied AEDs likely
provide our best interpretation of what to expect regarding dosing to be influenced by the inhibitory effects of fluoxetine or flu-
changes in the average patient after combining AEDs with new ADs voxamine are those dependent on CYP2C9 or CYP2C19 for their
(Table 4), combining new ADs with AEDs (Table 4) and combining metabolism, including clobazam, lacosamide, phenobarbital, or
AEDs with new APs (Table 5). These tables will provide some ori- primidone. Fluvoxamine may have also moderate inhibitory effects
84 E. Spina et al. / Pharmacological Research 106 (2016) 72–86

on AEDs dependent on CYP3A4 for their metabolism, such as car- [8] C. Johannesen Landmark, P. Patsalos, Drug interactions involving the new
bamazepine, felbamate, tiagabine, or zonisamide. Fluoxetine may second- and third-generation antiepileptic drugs, Expert Rev. Neurother. 10
(2010) 119–140.
take up to 2–3 months in an average patient (or even up to 6 months [9] G. Zaccara, E. Perucca, Interactions between antiepileptic drugs: and
in unusual patients) to reach maximal inhibitory effects on CYP3A4 between antiepileptic drugs and other drugs, Epileptic. Disord. 16 (2014)
due to the long half-life of norfluoxetine, so it may be important to 409–431.
[10] P.N. Patsalos, Drug interactions with newer antiepileptic drugs (AEDs)-part
remember that when co-prescribing fluoxetine with AEDs depen- 1: pharmacokinetic and pharmacodynamic interactions between AEDs, Clin.
dent on CYP3A4 such as carbamazepine, felbamate, tiagabine, or Pharmacokinet. 52 (2013) 927–966.
zonisamide. Table 4 shows that it may be safer to select ADs other [11] P.N. Patsalos, Drug interactions with newer antiepileptic drugs (AEDs)-part
2: pharmacokinetic and pharmacodynamic interactions between AEDs and
than fluoxetine or fluvoxamine when taking any of these AEDs.
drugs used to treat non-epilepsy disorders, Clin. Pharmacokinet. 52 (2013)
Isolated cases have suggested that fluoxetine and sertraline may 1045–1061.
influence lamotrigine TDM. Before we highlight these DIs for clin- [12] E. Spina, V. Santoro, C. D’Arrigo, Clinically relevant pharmacokinetic drug
interactions with second-generation antidepressants: an update, Clin. Ther.
icians, more definitive studies using high doses should establish
30 (2008) 1206–1227.
that sertraline inhibits lamotrigine metabolism or that fluoxetine [13] E. Spina, G. Trifirò, F. Caraci, Clinically significant drug interactions with
induces lamotrigine metabolism. newer antidepressants, CNS Drugs 26 (2012) 39–67.
[14] E. Spina, J. de Leon, Metabolic drug interactions with newer antipsychotics:
a comparative review, Basic Clin. Pharmacol. Toxicol. 100 (2007) 4–22.
Conflict of interest [15] W.K. Kennedy, M.W. Jann, E.C. Kutscher, Clinically significant drug
interactions with atypical antipsychotics, CNS Drugs 12 (2013) 1021–1048.
[16] M. Mula, F. Monaco, Antiepileptic-antipsychotic drug interactions: a critical
This work was not supported by any funding source. review of the evidence, Clin. Neuropharmacol. 25 (2002) 280–289.
In the past few years, Dr. Spina has participated in speak- [17] F.M.C. Besag, D. Berry, Interactions between antiepileptic and antipsychotic
ers/advisory boards and lectured supported by AstraZeneca, drugs, Drug Saf. 29 (2006) 95–118.
[18] M. Mula, Anticonvulsants-antidepressants pharmacokinetic drug
Bristol-Myers, Eli Lilly & Co, Janssen Pharmaceuticals, Lundbeck and interactions: the role of the CYP450 system in psychopharmacology, Curr.
Pfizer. Drug Metab. 8 (2008) 730–737.
In the past few years, Dr. Pisani has participated as speaker and [19] J. de Leon, V. Santoro, C. D’Arrigo, E. Spina, Interactions between
antiepileptics and second-generation antipsychotics, Exp. Opin. Drug Metab.
member of advisory boards supported by UCB Pharma, Eisai, Shire.
Toxicol. 8 (2012) 311–334.
Dr. de Leon personally develops his presentations for lecturing, [20] D. Italiano, E. Spina, J. de Leon, Pharmacokinetic and pharmacodynamic
has never lectured using any pharmaceutical or pharmacogenetic interactions between antiepileptics and antidepressants, Exp. Opin. Drug
Metab. Toxicol. 10 (2014) 1457–1489.
company presentations, and has never been a consultant for phar-
[21] K.L. Cozza, S.C. Armstrong, J.R. Oesterheld, Concise Guide to Drug Interaction
macogenetic or pharmaceutical companies. In the past, Dr. de Principles for Medical Practice: Cytochrome P450s, UGTs, P-Glycoproteins,
Leon received researcher-initiated grants from Eli Lilly (one ended 2nd ed., American Psychiatric Association, Washington, DC, 2003.
in 2003 and the other, as co-investigator, ended in 2007); from [22] J.H. Lin, Transporter-mediated drug interactions. Clinical implications and
in vitro assessment, Exp. Opin. Drug Metab. Toxicol. 3 (2007) 81–92.
Roche Molecular Systems, Inc. (ended in 2007); and, in a collabo- [23] C. Zhang, P. Kwan, Z. Zuo, L. Baum, The transport of antiepileptic drugs by
ration with Genomas, Inc., from the NIH Small Business Innovation P-glycoprotein, Adv. Drug Deliv. Rev. 64 (2012) 930–942.
Research program (ended in 2010). He has been on the advisory [24] F.E. O’Brien, T.G. Dinan, B.T. Griffin, J.F. Cryan, Interactions between
antidepressants and P-glycoprotein at the blood-brain barrier: clinical
boards of Bristol-Myers Squibb (2003/04) and AstraZeneca (2003). significance of in vitro and in vivo findings, Br. J. Pharmacol. 165 (2012)
Roche Molecular Systems supported one of his educational presen- 289–312.
tations, which was published in a peer-reviewed journal (2005). His [25] T. Moons, M. de Roo, S. Clees, G. Dom, Relationship between P-glycoprotein
and second generation antipsychotics, Pharmacogenomics 12 (2011)
lectures were supported once by Sandoz (1997), twice by Lundbeck 1193–1211.
(1999 and 1999), twice by Pfizer (2001 and 2001), three times by [26] Y. Akamine, N. Yasui-Furukori, I. Ieiri, T. Uno, Psychotropic drug–drug
Eli Lilly (2003, 2006, and 2006), twice by Janssen (2000 and 2006), interactions involving P-glycoprotein, CNS Drugs 26 (2012) 959–973.
[27] A.M. Kanner, The treatment of depressive disorders in epilepsy: what all
once by Bristol-Myers Squibb (2006), and seven times by Roche
neurologists should know, Epilepsia 54 (2013) 3–12.
Molecular Systems, Inc. (once in 2005 and six times in 2006). [28] P. Mehndiratta, M. Sajatovic, Treatments for patients with comorbid
epilepsy and depression: a systematic literature review, Epilepsy Behav. 28
(2013) 36–40.
Acknowledgment [29] L.N. Yatham, S.H. Kennedy, S.V. Parikh, et al., Canadian Network for Mood
and Anxiety Treatments (CANMAT) and International Society for Bipolar
The authors acknowledge Lorraine Maw, M.A., at the Mental Disorders (ISBD) collaborative update of CANMAT guidelines for the
management of patients with bipolar disorder: update 2013, Bipolar Disord.
Health Research Center at Eastern State Hospital, Lexington, KY, 15 (2013) 1–44.
who helped in editing the article. [30] K. Rickels, T.M. Shiovitz, T.S. Ramey, et al., Adjunctive therapy with
pregabalin in generalized anxiety disorder patients with partial response to
SSRI or SNRI treatment, Int. Clin. Psychopharmacol. 27 (2012) 142–150.
References [31] L.E. Chaparro, P.J. Wiffen, R.A. Moore, I. Gilron, Combination
pharmacotherapy for the treatment of neuropathic pain in adults, Cochrane
[1] E. Spina, D. Italiano, Drug interactions, in: S. Shorvon, E. Perucca, J. Engel Database Syst. Rev. 7 (2012) CD008943.
(Eds.), Treatment of Epilepsy, 4th ed., Wiley-Blackwell Publishing Ltd., [32] I. Gilron, T.S. Jensen, A.H. Dickenson, Combination pharmacotherapy for
Oxford, 2015, pp. 344–359. management of chronic pain: from bench to bedside, Lancet Neurol. 12
[2] E. Perucca, Clinically relevant drug interactions with antiepileptic drugs, Br. (2013) 1084–1095.
J. Clin. Pharmacol. 61 (2006) 246–255. [33] W.H. Greb, G. Buscher, H.D.H.D. Dierdorf, et al., The effect of liver enzyme
[3] E. Spina, E. Perucca, Clinical significance of pharmacokinetic interactions inhibition by cimetidine and enzyme induction by phenobarbitone on the
between antiepileptic and psychotropic drugs, Epilepsia 43 (Suppl. 2) (2002) pharmacokinetics of paroxetine, Acta Psychiatry Scand. 350 (Suppl) (1989)
37–44. 95–98.
[4] S.M. Stahl, Antidepressants, in: S.M. Stahl (Ed.), Stahl’s Essential [34] B.B. Andersen, M. Mikkelsen, A. Vesterager, et al., No influence of the
Psychopharmacology, 4th ed., Cambridge University Press, New York, 2013, antidepressant paroxetine on carbamazepine valproate and phenytoin,
pp. 284–369. Epilepsy Res. 10 (1991) 201–204.
[5] S.M. Stahl, Antipsychotic agents, in: S.M. Stahl (Ed.), Stahl’s Essential [35] A. Kahn, M.U. Shad, S.H. Preskhorn, Lack of sertraline efficacy probably due
Psychopharmacology, 4th ed., Cambridge University Press, New York, 2013, to an interaction with carbamazepine, J. Clin. Psychiatry 61 (2000) 526–527.
pp. 129–236. [36] M. Pihlsgard, E. Eliasson, Significant reduction of sertraline plasma levels by
[6] P.N. Patsalos, E. Perucca, Clinically important drug interactions in epilepsy: carbamazepine and phenytoin, Eur. J. Clin. Pharmacol. 57 (2002) 915–916.
general features and interactions between antiepileptic drugs, Lancet [37] L. Steinacher, P. Vandel, D.F. Zullino, et al., Carbamazepine augmentation in
Neurol. 2 (2003) 347–356. depressive patients non-responding to citalopram: a pharmacokinetic and
[7] P.N. Patsalos, E. Perucca, Clinically important drug interactions in epilepsy: clinical pilot study, Eur. Neuropsychopharmacol. 12 (2002) 255–260.
interactions between antiepileptic drugs and other drugs, Lancet Neurol. 2
(2003) 473–481.
E. Spina et al. / Pharmacological Research 106 (2016) 72–86 85

[38] C. Puozzo, B.E. Leonard, Pharmacokinetics of milnacipran in comparison [71] K. Linnet, O.V. Olesen, Free and glucuronidated olanzapine serum
with other antidepressants, Int. Clin. Psychopharmacol. 11 (Suppl. 4) (1996) concentrations in psychiatric patients: influence of carbamazepine
15–27. comedication, Ther. Drug Monit. 24 (2002) 512–517.
[39] J. Sitsen, F. Maris, C. Timmer, Drug-drug interaction studies with [72] E. Skogh, M. Reis, M. Dahl, et al., Therapeutic drug monitoring data on
mirtazapine and carbamazepine in healthy male subjects, Eur. J. Drug olanzapine and its N-demethyl metabolite in the naturalistic clinical setting,
Metab. Pharmacokinet. 26 (2001) 109–121. Ther. Drug Monit. 24 (2002) 518–526.
[40] E. Spaans, M.W. van den Heuvel, P.G. Schnabel, et al., Concomitant use of [73] S. Botts, F.J. Diaz, V. Santoro, et al., Estimating the effects of co-medications
mirtazapine and phenytoin: a drug–drug interactions study in healthy male on plasma olanzapine concentrations by using a mixed model, Progr. Neuro
subjects, Eur. J. Clin. Pharmacol. 58 (2002) 423–429. Psychopharmacol. Biol. Psychiatry 32 (2008) 1453–1458.
[41] T.A. Ketter, J.B. Jenkins, D.H. Schroeder, et al., Carbamazepine but not [74] T. Haslemo, K. Olsen, H. Lunde, E. Molden, Valproic acid significantly lowers
valproate induces bupropion metabolism, J. Clin. Psychopharmacol. 15 serum concentrations of olanzapine. An interaction effect comparable with
(1995) 327–333. smoking, Ther. Drug Monit. 34 (2012) 512–517.
[42] A. Helland, O. Spigset, Low serum concentrations of reboxetine in 2 patients [75] S.W. Grimm, N.M. Richtand, H.R. Winter, et al., Effects of cytochrome P450
treated with CYP3A4 inducers, J. Clin. Psychopharmacol. 27 (2007) 308–310. 3A modulators ketoconazole and carbamazepine on quetiapine
[43] J. de Leon, False-negative studies may systematically contaminate the pharmacokinetics, Br. J. Clin. Pharmacol. 61 (2006) 58–66.
literature on the effects of inducers in neuropsychopharmacology. Part II: [76] J. Hasselstrom, K. Linnet, Quetiapine serum concentrations in psychiatric
focus on bipolar disorder, J. Clin. Psychopharmacol. 34 (2014) 291–296. patients, Ther. Drug Monit. 26 (2004) 486–491.
[44] S. Unterecker, A. Reif, S. Hempel, et al., Interaction of valproic acid and the [77] I. Castberg, E. Skogvoll, O. Spigset, Quetiapine and drug interactions:
antidepressants doxepin and venlafaxine: analysis of therapeutic drug evidence from a routine therapeutic drug monitoring service, J. Clin.
monitoring data under naturalistic conditions, Int. Clin. Psychopharmacol. Psychiatry 68 (2007) 1540–1545.
29 (2014) 206–211. [78] V. Santoro, C. D’Arrigo, G. Migliardi, et al., Therapeutic drug monitoring of
[45] P. Jalil, Toxic reaction following the combined administration of fluoxetine quetiapine: effect of coadministration with antiepileptic drugs in patients
and phenytoin: two case reports, J. Neurol. Neurosurg. Psychiatry 55 (1992) with psychiatric disorders, Open Clin. Chem. J. 1 (2008) 17–21.
412–413. [79] M. Wittman, H. Hauser, A. Köstlbacher, et al., Individual clearance and
[46] J. Darley, Interaction between phenytoin and fluoxetine, Seizure 3 (1994) therapeutic drug monitoring of quetiapine in clinical practice,
151–152. Neuroendocrinol. Lett. 31 (2010) 203–207.
[47] D.J. Woods, D.M. Coulter, P. Pillans, Interaction of phenytoin and fluoxetine, [80] T. Nicki-Jockschat, M. Paulzen, F. Schneider, et al., Drug interaction can lead
NZ Med. J. 107 (1994) 19. to undetectable serum concentrations of quetiapine in the presence of
[48] R. Sovner, J.M. Davis, A potential drug interaction between fluoxetine and carbamazepine, Clin. Neuropharmacol. 32 (2009) 5.
valproic acid, J. Clin. Psychopharmacol. 11 (1991) 389. [81] J. de Leon, J. Bork, Risperidone and cytochrome P450 3A, J. Clin. Psychiatry
[49] S. Cruz-Flores, G.R. Hayat, W. Mirza, Valproic acid toxicity with fluoxetine 58 (1997) 450.
therapy, MO Med. 92 (1995) 296–297. [82] E. Spina, M.G. Scordo, A. Avenoso, et al., Adverse drug interaction between
[50] M.I. Lucena, E. Blanco, M.A. Corrales, M.L. Berthier, Interaction of fluoxetine risperidone and carbamazepine in a patient with chronic schizophrenia and
and valproic acid, Am. J. Psychiatry 155 (1998) 575. deficient CYP2D6, J. Clin. Psychopharmacol. 21 (2001) 108–109.
[51] H.J. Pearson, Interaction of fluoxetine with carbamazepine, J. Clin. Psychiatry [83] E. Spina, A. Avenoso, G. Facciolà, et al., Plasma concentrations of risperidone
51 (1990) 126. and 9-hydroxy-risperidone effect of comedication with carbamazepine or
[52] S.R. Grimsley, M.W. Jann, J.G. Carter, et al., Increased carbamazepine plasma valproate Ther, Drug Monit. 22 (2000) 481–485.
concentrations after fluoxetine coadmnistration, Clin. Pharmacol. Ther. 50 [84] S. Ono, K. Mihara, A. Suzuki, et al., Significant pharmacokinetic interaction
(1991) 10–15. between risperidone and carbamazepine: its relationship with CYP2D6
[53] E. Spina, A. Avenoso, A.M. Pollicino, et al., Carbamazepine coadministration genotypes, Psychopharmacology 162 (2002) 50–54.
with fluoxetine or fluvoxamine, Ther. Drug Monit. 15 (1993) 247–250. [85] N. Yasui-Furukori, K. Kubo, M. Ishioka, et al., Interaction between
[54] A. Reimers, E. Skogvoll, J.K. Sund, O. Spigset, Drug interactions between paliperidone and carbamazepine, Ther. Drug Monit. 35 (2013) 649–652.
lamotrigine and psychoactive drugs: evidence from a therapeutic drug [86] V. Kerbusch-Herben, A. Cleton, J. Berwaets, et al., Effect of carbamazepine on
monitoring service, J. Clin. Psychopharmacol. 25 (2005) 342–348. the pharmacokinetics of paliperidone extended-release tablets at steady
[55] J. Fritze, B. Unsorg, M. Lanczik, Interaction between carbamazepine and state, Clin. Pharmacol. Drug Dev. 3 (2014) 1–7.
fluvoxamine, Acta Psychiatry Scand. 84 (1991) 583–584. [87] J.J. Miceli, R.J. Anziano, L. Robarge, et al., The effect of carbamazepine on the
[56] P. Bonnet, S. Vandel, S. Nezelof, et al., Carbamazepine, fluvoxamine. Is there steady-state pharmacokinetics of ziprasidone in healthy volunteers, Br. J.
a pharmacokinetic interaction? Therapie 47 (1992) 165. Clin. Pharmacol. 49 (2000) 65S–70S.
[57] K. Mamiya, K. Kojima, E. Yukawa, et al., Phenytoin intoxication induced by [88] L. Citrome, J.P. Macher, D.E. Salazar, et al., Pharmacokinetics of aripiprazole
fluvoxamine, Ther. Drug Monit. 23 (2001) 75–77. and concomitant carbamazepine, J. Clin. Psychopharmacol. 27 (2007)
[58] W.G. Rapeport, S.A. Williams, D.C. Muirhead, et al., Absence of 279–283.
sertraline-mediated effect of the pharmacokinetics and pharmacodynamics [89] A. Nakamura, K. Mihara, G. Nagai, et al., Pharmacokinetic and
of carbamazepine, J. Clin. Psychiatry 57 (Suppl. 1) (1996) 20–23. pharmacodynamic interactions between carbamazepine and aripiprazole in
[59] W.G. Rapeport, D.C. Muirhead, S.A. Williams, et al., Absence of effect of patients with schizophrenia, Ther. Drug Monit. 31 (2009) 575–578.
sertraline on the pharmacokinetics and pharmacodynamics of phenytoin, J. [90] I. Castberg, O. Spigset, Effect of comedication on the serum level of
Clin. Psychiatry 57 (Suppl. 1) (1996) 24–28. aripiprazole: evidence from a routine therapeutic drug monitoring service,
[60] T.R. Berigan, J. Harazin, A sertraline/valproic acid drug interaction, Int. J. Pharmacopsychiatry 40 (2007) 107–110.
Psychiatr. Clin. Pract. 3 (1999) 287–288. [91] D.D. Miller, Effect of phenytoin on plasma clozapine concentrations in two
[61] K.R. Kaufman, R. Gerner, Lamotrigine toxicity secondary to sertraline, patients, J. Clin. Psychiatry 52 (1991) 23–25.
Seizure 7 (1998) 163–165. [92] H.Y. Lane, K.P. Su, W.H. Chang, et al., Elevated plasma clozapine
[62] J. Christensen, A.P. Sandgaard, P. Sidenius, et al., Lack of interaction between concentrations after phenobarbital discontinuation, J. Clin. Psychiatry 59
sertraline and lamotrigine in psychiatric patients: a retrospective study, (1998) 131–133.
Pharmacopsychiatry 45 (2012) 119–121. [93] G. Facciolà, A. Avenoso, E. Spina, E. Perucca, Inducing effect of phenobarbital
[63] S.E. Moller, F. Larsen, A.Z. Khan, P.E. Rolan, Lack of effect of citalopram on the on clozapine metabolism in patients with chronic schizophrenia, Ther. Drug
steady-state pharmacokinetics of carbamazepine in healthy male subjects, J. Monit. 20 (1998) 628–630.
Clin. Psychopharmacol. 21 (2001) 493–499. [94] Y.W. Wong, C. Yeh, P.T. Thyrum, The effects of concomitant phenytoin
[64] A. Sanchez-Romero, A. Mayordomo-Aranda, R. Garcia-Delgado, J.A. administration on the steady-state pharmacokinetics of quetiapine, J. Clin.
Duran-Quintana, Probable interaction between trazodone and Psychopharmacol. 21 (2001) 89–93.
carbamazepine, Pharmacopsychiatry 44 (2011) 158–159. [95] F. Centorrino, R.J. Baldessarini, J. Kando, et al., Serum concentrations of
[65] A.P. Popli, J. Tanquary, V. Lamparella, P.S. Masand, Bupropion and clozapine and its metabolites: effects of cotreatment with fluoxetine or
anticonvulsant drug interactions, Ann. Clin. Psychiatry 7 (1995) 99–101. valproate, Am. J. Psychiatry 151 (1994) 123–125.
[66] J. Odishaw, C. Chen, Effects of steady-state bupropion on the [96] G. Facciolà, A. Avenoso, M.G. Scordo, et al., Small effects of valproic acid on
pharmacokinetics of lamotrigine in healthy subjects, Pharmacotherapy 20 the plasma concentrations of clozapine and its major metabolites in patients
(2000) 1448–1453. with schizophrenic or affective disorders, Ther. Drug Monit. 21 (1999)
[67] M. Jerling, L. Lindstrom, U. Bondesson, L. Bertilsson, Fluvoxamine inhibition 341–345.
and carbamazepine induction of the metabolism of clozapine: evidence [97] J.O. Wong, S.P. Leung, T. Mak, et al., Plasma clozapine levels and clinical
from a therapeutic drug monitoring service, Ther. Drug Monit. 16 (1994) response in treatment-refractory Chinese schizophrenic patients, Prog.
368–374. Neuro Psychopharmacol. Biol. Psychiatry 30 (2006) 251–264.
[68] J. Tiihonen, H. Bartiainen, P. Hakola, Carbamazepine-induced changes in [98] A.P. Rajkumar, B. Poonkuzhali, A. Kuruvilla, et al., Clinical predictors of
plasma levels of neuroleptics, Pharmacopsychiatry 28 (1995) 26–28. serum clozapine levels in patients with treatment-resistant schizophrenia,
[69] R.A. Lucas, D.J. Gilfillan, R.F. Bergstrom, A pharmacokinetic interaction Int. Clin. Psychopharmacol. 28 (2013) 50–56.
between carbamazepine and olanzapine: observations on possible [99] P. Finley, D. Warner, Potential impact of valproic acid therapy on clozapine
mechanisms, Eur. J. Clin. Pharmacol. 54 (1998) 639–643. disposition, Biol. Psychiatry 36 (1994) 487–488.
[70] O.V. Olesen, K. Linnet, Olanzapine serum concentrations in psychiatric [100] F.J. Diaz, V. Santoro, E. Spina, et al., Estimating the size of the effects of
patients given standard doses: the influence of comedication, Ther. Drug co-medications on plasma clozapine concentrations using a model that
Monit. 21 (1999) 87–90.
86 E. Spina et al. / Pharmacological Research 106 (2016) 72–86

controls for clozapine doses and confounding variables, [116] J. Sidhu, S. Job, J. Bullman, et al., Pharmacokinetics and tolerability of
Pharmacopsychiatry 41 (2008) 81–91. lamotrigine and olanzapine coadministered to healthy subjects, Br. J Clin.
[101] F.J. Diaz, C.B. Eap, N. Ansermot, et al., Can valproic acid be an inducer of Pharmacol. 61 (2006) 420–426.
clozapine metabolism, Pharmacopsychiatry 47 (2014) 89–96. [117] M.L. Andersson, L. Bjorkhem-Bergman, J.D. Lindh, Possible
[102] A. Riesselman, B. Strobl, A.T. Cooley, J. de Leon, A case report that suggested drug-druginteraction between quetiapine and lamotrigine–evidence from a
that aspirin’s effects on valproic acid metabolism may contribute to valproic Swedish TDM database, Br. J. Clin. Pharmacol. 72 (2011) 153–156.
acid’s inducer effects on clozapine metabolism, J. Clin. Psychopharmacol. 33 [118] S.D. Bienentreu, K.T.H. Kronmuller, Increase in risperidone plasma level with
(2013) 812–814. lamotrigine, Am. J. Psychiatry 162 (2005) 811–812.
[103] N. Chopra, J. de Leon, Clozapine-induced myocarditis may be associated [119] R.B. Waade, H. Christensen, I. Rudberg, et al., Influence of comedication on
with rapid titration: a case report verified with autopsy, Int. J. Psychiatry serum concentrations of aripiprazole and dehydroaripiprazole, Ther. Drug
Med. 51 (2016) 104–115. Monit. 31 (2009) 233–238.
[104] M. Gex-Fabry, A.E. Balant-Gorgia, L. Balant, Therapeutic drug monitoring of [120] M.R. Muscatello, M. Pacetti, M. Cacciola, et al., Plasma concentrations of
olanzapine: the combined effect of age gender, smoking, and comedication, risperidone and olanzapine during coadministration with oxcarbazepine,
Ther. Drug Monit. 25 (2003) 46–53. Epilepsia 46 (2005) 771–774.
[105] N. Bergemann, K.R. Kress, F. Abu-Tair, et al., Valproate lowers plasma [121] I.R. McGrane, J.G. Loveland, H.J. Zaluski, K.D. Foster, Serum quetiapine
concentration of olanzapine, J. Clin. Psychopharmacol. 26 (2006) 432–434. concentration changes with concomitant oxcarbazepine therapy in a boy
[106] E. Spina, C. D’Arrigo, V. Santoro, et al., Effect of valproate on olanzapine with autism spectrum disorder, J. Child Adolesc. Psychopharmacol. 25
plasma concentrations in patients with bipolar or schizoaffective disorder, (2015) 729–730.
Ther. Drug Monit. 31 (2009) 758–763. [122] M. Bialer, D.R. Doose, B. Murthy, et al., Pharmacokinetic interactions of
[107] W. Aichhorn, J. Marksteiner, T. Walch, et al., Influence of age gender, body topiramate, Clin. Pharmacokinet. 43 (2004) 763–780.
weight and valproate co-medication on quetiapine plasma concentrations, [123] G. Migliardi, C. D’Arrigo, V. Santoro, et al., Effect of topiramate on plasma
Int. Clin. Psychopharmacol. 21 (2006) 81–85. concentrations of clozapine olanzapine, risperidone, and quetiapine in
[108] H.R. Winter, C.L. DeVane, C. Figueroa, et al., Open-label steady-state patients with psychotic disorders, Clin. Neuropharmacol. 30 (2007)
pharmacokinetic drug interaction study on co-administered quetiapine 107–113.
fumarate and divalproex sodium in patients with schizophrenia [124] M. Mula, F. Monaco, Carbamazepine-risperidone interaction in patients with
schizoaffective disorder, or bipolar disorder, Hum. Psychopharmacol. Clin. epilepsy, Clin. Neuropharmacol. 25 (2002) 97–100.
Exp. 22 (2007) 469–476. [125] B. Fitzgerald, A. Okos, Elevation of carbamazepine-10:11-epoxide by
[109] A. Ravindran, P. Silverstone, D. Lacroix, et al., Risperidone does not affect quetiapine, Pharmacother. 22 (2002) 1500–1503.
steady-state pharmacokinetics of divalproex sodium inpatients with bipolar [126] D.W. Boulton, G.D. Kollia, S. Mallikaarjun, D.M. Kornhauser, Lack of a
disorder, Clin. Pharmacokinet. 43 (2004) 733–740. pharmacokinetic drug–drug interaction between lithium andvalproate
[110] L. Citrome, R. Josiassen, N. Bark, et al., Pharmacokinetics of aripiprazole and when co-administered with aripiprazole, J. Clin. Pharm. Ther. 37 (2012)
concomitant lithium and valproate, J. Clin. Pharmacol. 45 (2005) 89–93. 565–570.
[111] M.G. Gerrits, R. de Greef, P. Dogterom, et al., Valproate reduces the [127] A. Reimers, E. Skogvoll, J.K. Sund, et al., Drug interaction between
glucuronidation of asenapine without affecting asenapine plasma lamotrigine and psychoactive drugs, J. Clin. Psychopharmacol. 25 (2005)
concentrations, J. Clin. Pharmacol. 52 (2012) 757–765. 342–348.
[112] M. Kossen, J.P. Selten, R.S. Kahn, Elevated clozapine plasma level with [128] F.C. Schieber, D.W. Boulton, A.H. Balch, et al., A non-randomized study to
lamotrigine, Am. J. Psychiatry 158 (2001) 1930. investigate the effects of the atypical antipsychotic aripiprazole on the
[113] J. Tiihonen, T. Hallikainen, O.P. Ryynanen, et al., Lamotrigine in steady-state pharmacokinetics of lamotrigine in patients with bipolar I
treatment-resistant schizophrenia: a randomized placebo-controlled disorder, Hum. Psychopharmacol. Clin. Exp. 24 (2009) 145–152.
crossover trial, Biol. Psychiatry 54 (2003) 1241–1248. [129] J. de Leon, E. Spina, F.J. Diaz, Pharmacokinetic drug interaction studies must
[114] E. Spina, C. D’Arrigo, G. Migliardi, et al., Effect of adjunctive lamotrigine consider pharmacological heterogeneity use of repeated dosing, and
treatment on the plasma concentrations of clozapine: risperidone and translation into a message understandable to practicing clinicians, J. Clin.
olanzapine in patients with schizophrenia or bipolar disorder, Ther. Drug Psychopharmacol. 29 (2009) 201–205.
Monit. 28 (2006) 599–602.
[115] M.W. Jann, Y.Y. Hon, S.A. Shamsi, et al., Lack of pharmacokinetic interaction
between lamotrigine and olanzapine in healthy volunteers,
Pharmacotherapy 26 (2006) 627–633.

You might also like