throbber
Review
`
`Click here for more articles from the symposium
`
`doi: 10.1111/joim.12317
`
`Polymorphism in CYP2D6 and CYP2C19, members of the
`cytochrome P450 mixed-function oxidase system, in the
`metabolism of psychotropic drugs
`
`J. Stingl1 & R. Viviani2
`From the 1Center for Translational Medicine, University of Bonn Medical School, Bonn; and 2Department of Psychiatry and Psychotherapy III,
`University of Ulm, Ulm, Germany
`
`Abstract. Stingl J, Viviani R (University of Bonn
`Medical Faculty, Bonn; University of Ulm, Ulm;
`Germany).
`Polymorphism in CYP2D6
`and
`CYP2C19, members of
`the cytochrome P450
`mixed-function
`oxidase
`system,
`in
`the
`metabolism of psychotropic drugs
`(Review).
`J Intern Med 2015; 277: 167–177.
`
`Numerous studies in the field of psychopharmaco-
`logical treatment have investigated the possible
`contribution of genetic variability between individ-
`uals to differences in drug efficacy and safety,
`motivated by the wide individual variation in
`treatment response. Genomewide analyses have
`been conducted in several large-scale studies on
`antidepressant drug response. However, no con-
`sistent findings have emerged from these studies.
`In a recent meta-analysis of genomewide data from
`the three studies capturing common variation for
`association with symptomatic improvement and
`remission revealed the absence of any strong
`genetic association and failed to replicate results
`of individual studies in the pooled data. However,
`
`there are good reasons to consider the possible
`importance of pharmacogenetic variants sepa-
`rately. These variants explain a large portion of
`the manifold variability in individual drug metab-
`olism. More than 20 psychotropic drugs have now
`been relabelled by the FDA adding information on
`polymorphic drug metabolism and therapeutic
`recommendations. Furthermore, dose recommen-
`dations for polymorphisms in drug metabolizing
`enzymes, first
`and foremost CYP2D6 and
`CYP2C19, have been issued with the advice to
`reduce the dosage in poor metabolizers to 50% or
`less (in eight cases), or to choose an alternative
`treatment. Beside the well-described role in hepatic
`drug metabolism,
`these
`enzymes
`are
`also
`expressed in the brain and play a role in biotrans-
`formation of endogenous substrates. These poly-
`morphisms may
`therefore modulate
`brain
`metabolism and affect the function of the neural
`substrates of cognition and emotion.
`
`Keywords: antidepressant drugs, CYP2C19, CYP2D6,
`drug metabolism, pharmacogenetics.
`
`Pharmacogenomics of antidepressant drug response: the
`genomewide association study approach
`
`Psychotropic medications belong to the most
`frequently prescribed drugs with more than
`100Mio daily drug doses in Germany in long-term
`therapies [1]. Hence,
`the fact
`that
`individual
`response to the same drug given in the same dose
`varies considerably has large practical
`implica-
`tions.
`In antidepressant treatment, only about
`one-third of patients achieve sufficient long-term
`symptomatic remission with their first-choice
`antidepressant, whilst the remaining 67% do not
`respond and are subsequently switched to different
`antidepressant drugs with the consequence of a
`prolongation of the duration of illness [2].
`
`Evidence for the hypothesis that genetic variation
`may contribute to the variability of drug response
`has been intensively sought. Several large-scale
`genetic association studies on antidepressant drug
`response have been recently performed using the
`genomewide association approach [3–5]. However,
`a consistent picture of replicated findings has so
`far failed to emerge.
`
`In a recent meta-analysis, the results from these
`three studies were combined with the aim to
`increase the power for the detection of effects [6].
`A broader analysis was conducted including all
`patients to reveal general genetic patterns of
`remission, whilst a narrower analysis in patients
`focused on selective serotonin reuptake inhibitors
`
`ª 2014 The Association for the Publication of the Journal of Internal Medicine
`
`167
`
`Vanda Exhibit 2042 - Page 1
`
`

`

`J. Stingl & R. Viviani
`
`Review: Pharmacogenetics of psychotropic drugs
`
`(SSRIs), especially escitalopram or citalopram.
`Using a genetic imputation technique, genome-
`wide data pooled from the three studies capturing
`common variation for association with symptom-
`atic improvement and remission were analysed.
`The results revealed the lack of strong genetic
`associations, and failed to replicate the single
`study results in the pooled analysis. The broader
`analysis in the entire sample resulted in one
`genomewide effect of a SNP located in the intronic
`region of the myosin X (MYO10) gene at 5p15.1,
`but the result could not be validated in confir-
`matory follow-up genotyping because of
`the
`absence of this effect in the STAR*D study [6].
`The narrow analysis focusing on SSRI-treated
`individuals identified a variant associated with
`early SSRI response (within 2 weeks of
`treat-
`ment), a SNP in an intergenic region on chromo-
`some 5. This association awaits replication in
`future studies [6].
`
`The failure to replicate findings and the poor
`predictive value of genetic variants in genomewide
`association studies raises the issue of the real
`prospects of success of approaches that wager on
`exploratory analyses to compensate for our limited
`current understanding of how neurobiological
`mechanisms and molecular changes contribute to
`the genesis of psychiatric disorders. This issue will
`likely take a prominent position in research in
`psychopharmacology and treatment of psychiatric
`disorders in the years to come.
`
`Pharmacogenetics: Psychotropic drugs and polymorphic drug
`metabolism
`
`A different approach focuses on the genetic vari-
`ability in drug metabolizing enzymes (DMEs) [7].
`These genetic polymorphisms have large effects,
`extensively documented in studies of individual
`differences in drug clearance affecting therapy
`outcome and safety [8].
`
`DME phenotypes, as predicted from genotypes,
`may be classified into four major groups:
`
`1 poor metabolizers (PM phenotype) are charac-
`terized by a complete lack of enzyme activity (two
`defective alleles);
`
`2 intermediate metabolizers (IM phenotype) are
`carriers of either one defective allele or two partially
`functional alleles;
`
`168 ª 2014 The Association for the Publication of the Journal of Internal Medicine
`Journal of Internal Medicine, 2015, 277; 167–177
`
`3 extensive metabolizers (EM phenotype) are car-
`riers of two functional alleles;
`
`4 ultrarapid metabolizers (UM phenotype) are car-
`riers of alleles leading to increased enzyme func-
`tion.
`
`As differences in plasma concentration due to
`individual variability in drug clearance in these
`phenotypes can be more than 10-fold, the possi-
`bility of delivering ‘personalized medicine’ through
`dose adjustments based on pharmacokinetic data
`has a sound rationale [9–11]. Most psychiatric
`drugs are extensively metabolized by these poly-
`morphic DMEs, with CYP2D6 being involved in the
`metabolism of approximately half of the commonly
`prescribed psychotropic drugs [12].
`
`dose-adjustment
`pharmacokinetic-based
`The
`approach is now being integrated with a formal
`assessment of the degree of evidence justifying a
`clinical recommendation for specific drug–geno-
`type pairs (Clinical Pharmacogenetics Implemen-
`tation consortium, CPIC) [10, 13]. Evidence-based
`dosing guidelines have now been issued for 27
`drugs and made available in the PharmGKB data-
`base [14]. Of all genes involved in these evalua-
`tions, CYP2C19 and CYP2D6 have been shown to
`be the most important DMEs for dose adjustments
`of psychotropic drugs.
`
`This empirical evidence is starting to have an
`impact on regulatory agencies such as the FDA
`and the EMA. Information on the genetic polymor-
`phism in DMEs is now being inserted in drug
`labels, where it is given in the respective sections of
`the product information (Dosage and Administra-
`tion, Warnings and Precautions, Drug Interactions,
`Clinical Pharmacology) [15]. To date, more than
`150 drug labels have been updated with pharma-
`cogenetic information by the FDA and more than
`70 by the EMA [16] (http://www.pharmgkb.org/
`view/drug-labels.do), alerting physicians
`that
`genetic factors may influence therapeutic outcome
`or adverse reactions. Of these pharmacogenetic
`labels, 24 involve drugs used in psychiatry. In 23
`cases, alertness is recommended due to major
`metabolism via CYP2D6, whilst four cases involve
`CYP2C19. For most drugs in psychiatric use, there
`are no explicit dose adjustments for the different
`phenotype groups, but rather general recommen-
`dations to watch out for and adopt a cautious
`stance about drug interactions. In Table 1, we have
`
`Vanda Exhibit 2042 - Page 2
`
`

`

`J. Stingl & R. Viviani
`
`Review: Pharmacogenetics of psychotropic drugs
`
`Table 1 Pharmacokinetic based dose adjustments for psychotropic drugs for which pharmacogenetic information is provided
`in the drug label
`
`PK differences caused by
`
`genotype corresponding to
`
`Pharmacokinetic
`
`Clinical-based
`
`dose adjustments
`
`implications (toxicity
`
`evidence
`
`Substance
`
`DME
`
`PM
`
`IM
`
`EM
`
`UM
`
`or efficacy)
`
`studies
`
`Action to be
`
`undertaken
`
`Antidepressants
`
`Amitriptyline
`
`CYP2D6
`
`CYP2C19 70% 87% 105
`
`67% 90% 114% 138%a Toxicity/side effect
`risk in PM, risk for
`
`141%
`
`therapeutic failure in
`
`efficacy and
`
`UMs
`
`toxicity
`
`Limited
`
`Monitor plasma
`
`evidence for
`
`concentrations
`(P+M),
`Consider dose
`
`adjustment
`
`Clomipramine CYP2D6
`
`55% 87% 119% 152% Toxicity/side effect
`
`Limited
`
`Monitor plasma
`
`CYP2C19 69% 87% 106% 125%
`
`risk in PM, risk for
`
`evidence for
`
`therapeutic failure in
`
`efficacy and
`
`UMs
`
`toxicity
`
`concentrations
`(P+M),
`Consider dose
`
`adjustment
`
`Desipramine
`
`CYP2D6
`
`25% 76% 136% 207% Toxicity/side effect
`
`Limited
`
`Monitor plasma
`
`risk in PM, risk for
`
`evidence for
`
`therapeutic failure in
`
`efficacy and
`
`UMs
`
`toxicity
`
`concentrations
`(P+M),
`Consider dose
`
`adjustment
`
`Doxepin
`
`CYP2D6
`
`34% 77% 131% 204% Toxicity/side effect
`
`Limited
`
`Monitor plasma
`
`risk in PM, risk for
`
`evidence for
`
`therapeutic failure in
`
`efficacy and
`
`UMs
`
`toxicity
`
`concentrations
`(P+M),
`Consider dose
`
`adjustment
`
`Imipramine
`
`CYP2D6
`
`28% 79% 131% 183% Toxicity/side effect
`
`Limited
`
`Monitor plasma
`
`CYP2C19 77% 86% 106% 128%
`
`risk in PM, risk for
`
`evidence for
`
`therapeutic failure in
`
`efficacy and
`
`UMs
`
`toxicity
`
`concentrations
`(P+M),
`Consider dose
`
`adjustment
`
`Nortriptyline
`
`CYP2D6
`
`50% 72% 133% 195% Toxicity/side effect
`
`Limited
`
`Monitor plasma
`
`risk in PM, risk for
`
`evidence for
`
`therapeutic failure in
`
`efficacy and
`
`UMs
`
`toxicity
`
`concentrations
`(P+M),
`Consider dose
`
`adjustment
`
`Trimipramine CYP2D6
`
`59% 88% 118% 147% Toxicity/side effect
`
`Limited
`
`Monitor plasma
`
`CYP2C19 45% 76% 107% 154%
`
`risk in PM, risk for
`
`evidence for
`
`therapeutic failure in
`
`efficacy and
`
`UMs
`
`toxicity
`
`concentrations
`(P+M),
`Consider dose
`
`adjustment
`
`ª 2014 The Association for the Publication of the Journal of Internal Medicine
`Journal of Internal Medicine, 2015, 277; 167–177
`
`169
`
`Vanda Exhibit 2042 - Page 3
`
`

`

`J. Stingl & R. Viviani
`
`Review: Pharmacogenetics of psychotropic drugs
`
`Table 1 (Continued )
`
`PK differences caused by
`
`genotype corresponding to
`
`Pharmacokinetic
`
`Clinical-based
`
`dose adjustments
`
`implications (toxicity
`
`evidence
`
`Substance
`
`Citalopram
`
`DME
`
`UM
`EM
`IM
`PM
`or efficacy)
`CYP2C19 59% 87% 107% 130% Large therapeutic
`
`studies
`
`Action to be
`
`undertaken
`
`Conflicting
`
`Monitor plasma
`
`Fluoxetine
`
`CYP2D6
`
`–
`
`–
`
`–
`
`–
`
`range; no response
`
`evidence on
`
`concentrations,
`
`effect observed except
`
`response and
`
`Consider dose
`
`UMs carrying the risk
`
`toxicity
`
`adjustment
`
`of therapeutic failure
`
`Substrate and strong
`inhibitor, P+M equal
`for CYP2D6
`
`No clinical
`
`Alertness for drug
`
`evidence for
`
`interactions with
`
`important
`
`CYP2D6
`
`genotypes, no data on
`
`genotype
`
`substrates
`
`safety risk for PMs
`
`effect
`
`Fluvoxamine
`
`CYP2D6
`
`68% 89% 117% 147% Preliminary PK data
`
`Clinical
`
`Consider
`
`only
`
`evidence for
`
`monitoring
`
`response and
`
`plasma
`
`toxicity
`
`concentrations,
`
`Paroxetine
`
`CYP2D6
`
`51% 81% 125% 169% Substrate and
`
`No clinical
`
`Select alternative
`
`dose adjustments
`
`inhibitor
`
`evidence for
`
`drug in UMs,
`
`response,
`
`consider dose
`
`clinical
`
`adjustment in PM
`
`Venlafaxine
`
`CYP2D6
`
`77% 92% 109% 172% Active metabolites.
`
`evidence for
`
`adverse
`
`effects
`
`Clinical
`
`Select alternative
`
`Safety concern in PMs
`
`evidence for
`
`drug in PM, UM,
`
`response and
`
`and IM
`
`toxicity
`
`CYP2C19 (44% 89% 107% 124%) Preliminary PK data
`
`No clinical
`
`Consider dose
`
`only, Safety concern
`
`evidence
`
`adjustment
`
`in PMs
`
`Antipsychotics
`
`Perphenazine CYP2D6
`
`(45% 67% 116% 164%) Only preliminary PK
`
`No evidence on
`
`No
`
`data available
`
`toxicity
`
`recommendations
`
`Pimozide
`
`(CYP2D6)
`
`Only minor role of
`
`No clinical
`
`No actions
`
`polymorphic DMEs
`
`evidence data
`
`necessary
`
`Thioridazine
`
`CYP2D6
`
`42% 83% 125% 153% Only PK data available
`
`Limited
`
`Consider dose
`
`evidence on
`
`adjustment
`
`toxicity
`
`170 ª 2014 The Association for the Publication of the Journal of Internal Medicine
`Journal of Internal Medicine, 2015, 277; 167–177
`
`Vanda Exhibit 2042 - Page 4
`
`

`

`J. Stingl & R. Viviani
`
`Review: Pharmacogenetics of psychotropic drugs
`
`Table 1 (Continued )
`
`PK differences caused by
`
`genotype corresponding to
`
`Pharmacokinetic
`
`Clinical-based
`
`dose adjustments
`
`implications (toxicity
`
`evidence
`
`Substance
`
`DME
`
`Aripiprazole
`
`CYP2D6
`
`UM
`EM
`IM
`PM
`or efficacy)
`66% 90% 115% 139% Large therapeutic
`
`studies
`
`Limited
`
`Action to be
`
`undertaken
`
`Consider dose
`
`range; no response
`
`clinical
`
`adjustment in
`
`effect expected except
`
`evidence
`
`UMs
`
`UMs carrying the risk
`
`of therapeutic failure
`
`Clozapine
`
`(CYP1A2)
`
`Not metabolized by
`
`No clinical
`
`No actions
`
`Risperidone
`
`CYP2D6
`
`polymorphic DMEs
`
`evidence for
`
`necessary
`
`56% 88% 119% 146%a Safety concern in PMs,
`P+M equally effective
`across CYP2D6
`
`an effect
`
`Clinical
`
`Select alternative
`
`evidence on
`
`drug in PM, UM
`
`response and
`
`and IM
`
`genotypes
`
`toxicity
`
`Diazepam
`
`CYP2C19 (51% 81% 109% 138%) Preliminary PK data
`
`No clinical
`
`No
`
`Atomoxetine
`
`CYP2D6
`
`–
`
`–
`
`–
`
`–
`
`Preliminary PK data
`
`No clinical
`
`Standard dose in
`
`only
`
`evidence data
`
`recommendations
`
`only; insufficient data
`
`evidence data
`
`PM, IM; Be alert
`
`to allow calculation of
`
`dose adjustment
`
`to reduced
`
`efficacy or select
`
`alternative drug
`
`in UM
`
`aBased on sum of pharmacologically active parent drug and active metabolite.
`
`collected recommendations of pharmacokinetic
`dose adjustments for the psychotropic drugs in
`common use, and for which such labelling have
`been issued by the regulatory agencies (for more
`detailed information, see FDA website and [11]).
`
`The overall impact of dose adjustments on clinical
`decisions also depends on considerations about
`the clinical utility of equalizing pharmacokinetic
`differences. For example, at one extreme of the
`range of clinical consequences of DME polymor-
`phisms, inadequate exposure to a drug may con-
`stitute a risk to life due to nonresponse or toxicity.
`At the other extreme, imprecise dosing may have
`little or no clinical consequences, because of a wide
`therapeutic range. When the drug is safe and/or
`effective within a plasma concentration range,
`genetic information may be used together with
`therapeutic drug monitoring to ensure adequate
`treatment
`(a resource
`giving
`evidence-based
`
`plasma concentration ranges for psychotropic
`drugs is [17]). Evidence on the clinical relevance
`of DME polymorphism and inadequate dosing in
`antidepressant drug treatment comes from clinical
`data or epidemiological and safety data, showing
`and association of the CYP2D6 poor metabolizer
`phenotype with increased times to settle on the
`appropriate drug and more frequent drug switches
`during antidepressant treatment [18]. These data
`also suggest that poor metabolizers suffer more
`frequently from adverse drug reactions than exten-
`sive metabolizers and register longer hospital
`stays, whereas ultrarapid metabolizers have a
`higher risk of therapeutic failure [18–22].
`
`Expression of DMEs in the brain
`
`Several DMEs are expressed throughout the body,
`including the brain [23], suggesting that they may
`be metabolically active locally and play a role in the
`
`ª 2014 The Association for the Publication of the Journal of Internal Medicine
`Journal of Internal Medicine, 2015, 277; 167–177
`
`171
`
`Vanda Exhibit 2042 - Page 5
`
`

`

`J. Stingl & R. Viviani
`
`Review: Pharmacogenetics of psychotropic drugs
`
`regulation of physiological homoeostasis by bio-
`transformation of endogenous compounds [24].
`Brain-expressed DMEs are distributed to specific
`brain regions and different cell types [25–29]. Most
`studies have investigated CYP2D6, which is
`expressed in both neuronal and in glial cells [26,
`30–35]. In the brain, DME gene expression appears
`to be regulated differently than in the liver. Higher
`CYP2D6 protein levels have been reported in brain
`
`of smokers and alcoholics with unchanged hepatic
`enzyme levels [26, 36].
`
`Polymorphisms in CYP2C19 as well as CYP2D6
`may have effects on brain function with conse-
`quences on affect and affective disorders [37, 38].
`In man, there is evidence of a direct association of
`CYP2C19 polymorphism and depression [37].
`Interestingly, CYP2C19 seems to be expressed only
`
`Genotype
`
`Brain DME
`activity
`
`Brain endogenous
`substrate activity
`
`(a)
`
`Comedication/other
`DME substrates
`
`Liver DME
`activity
`
`Brain function
`
`Treatment success
`neurotoxicity
`behavioral effect
`
`Drug of
`interest/xenobiotic
`
`Blood concentration
`drug/xenobiotic
`
`Genotype
`
`Brain
`morphogenesis
`
`CYP2D6
`
`CYP2C19
`
`(b)
`
`Comedication/other
`DME substrates
`
`Liver DME
`activity
`
`Brain function
`
`Treatment success
`behavioral effect
`
`Drug of
`interest/xenobiotic
`
`Blood concentration
`drug/xenobiotic
`
`Legend:
`
`Induction/inhibition
`
`Fig. 1 Graph illustrating causal paths considered in the literature linking genotype variants, xenobiotic exposures, and
`effects with individual differences in treatment efficacy, cognitive function, vulnerability to neurotoxins or behavioural
`disorders. These causal paths need to be considered when inferring in observational studies the effect of genotype on
`behaviour or vulnerability to disorder [61]. The drug of interest here may also be any xenobiotic, such as a neurotoxin, or a
`substance of abuse, that is a substrate of the drug metabolizing enzyme in question. a, top: effects of genetic polymorphism
`in CYP2D6. This complex graph reflects the manifold effects due to the activity of CYP2D6 in the liver and in the brain, where
`it may simultaneously affect brain function and act on endogenous and exogenous substrate. b, bottom: effects of genetic
`polymorphism in CYP2C19. Current evidence suggests that the effect on brain function of CYP2C19 is mediated by
`morphological differences emerging during development.
`
`172 ª 2014 The Association for the Publication of the Journal of Internal Medicine
`Journal of Internal Medicine, 2015, 277; 167–177
`
`Vanda Exhibit 2042 - Page 6
`
`

`

`J. Stingl & R. Viviani
`
`Review: Pharmacogenetics of psychotropic drugs
`
`Fig. 2 With copyright permission from [51]. Top half: Regression of resting brain perfusion values on the CYP2D6 activity
`score over the whole brain. Left: hypothalamus and occipital/calcarine cortex; Centre: hippocampus and adjacent occipito/
`temporal cortex (lingual and fusiform gyrus); Right: thalamus and occipital/calcarine cortex. Bottom half: cerebral blood flow
`perfusion in the hypothalamus and occipital/calcarine region divided by CYP2D6 activity scores. Individuals with activity 0
`in this plot are poor metabolizers.
`
`during brain development in the foetus. However,
`in mice, this expression has morphological and
`functional effects on the brain in adult life [39]. A
`decreased hippocampal volume, an altered neuro-
`nal composition in the hippocampal dentate gyrus,
`and behaviour indicative of increased stress and
`anxiety based on four different tests were observed
`[39]. These results indicate that CYP2C19 metab-
`olizes critical compounds for brain development
`during foetal life, potentially affecting brain devel-
`opment. A number of independent studies have
`also associated the CYP2D6 polymorphism with
`the risk of suicide. Forensic studies originally
`reported an increased frequency of the ultrarapid
`
`metabolizer type in suicide victims [40–43]. Two
`further studies in depressives [38] and eating
`disorders [44] showed higher suicidality and sui-
`cidal ideation in carries of the ultrarapid metabo-
`lizer phenotype.
`
`No conclusive evidence identifies the possible
`endogenous substrates of CYP2D6 or other DMEs
`that may underlie these effects on brain function.
`Plausible candidates include monoamines, neu-
`rosteroids and endorphins [45, 46]. However, evi-
`dence on an effect of CYP2D6 genotype on
`serotonergic or dopaminergic function in vivo
`remains sparse [47, 48].
`
`ª 2014 The Association for the Publication of the Journal of Internal Medicine
`Journal of Internal Medicine, 2015, 277; 167–177
`
`173
`
`Vanda Exhibit 2042 - Page 7
`
`

`

`J. Stingl & R. Viviani
`
`Review: Pharmacogenetics of psychotropic drugs
`
`(a)
`
`PAROXETINE
`
`(b)
`
`BUPROPION
`
`(c)
`
`PAROXETINE
`vs. BUPROPION
`
`x = 5 mm
`
`x = 5 mm
`
`x = 5 mm
`
`(d)
`
`(e)
`
`(f)
`
`t
`
`−4.0
`
`−3.5
`
`−2.9
`
`−1.75
`1.75
`
`z = −30 mm
`
`z = −30 mm
`
`z = −30 mm
`
`Fig. 3 With copyright permission to be reproduced from
`[58]. Effects of paroxetine (a, d) and bupropion (b, e) versus
`placebo and of the comparison paroxetine versus bupro-
`pion (c, f) in the brainstem. In the paroxetine and bupro-
`pion versus placebo comparisons, light blue and magenta
`colours refer to decrements of perfusion under medication.
`In the comparison paroxetine versus bupropion, light blue
`and magenta colours are compatible with larger perfusion
`decrements under paroxetine, whilst yellow colour is
`compatible with larger perfusion decrements under bupro-
`pion. Threshold for visualization purposes: P < 0.005 (full
`colours) and P < 0.05 (light blue, yellow), uncorrected.
`
`Because of the potential simultaneous effect of
`CYP2D6 on the metabolism of both xenobiotics
`and endobiotics and its localization in the liver
`and in the brain, the overall effect of its poly-
`morphism can be due to a set of alternative or
`concurrent
`causal paths affecting
`individual
`treatment or behavioural effects (Fig. 1). An
`example is given by the possible association
`between the ultrarapid metabolizer phenotype
`and suicide. A possible, and perhaps most obvi-
`ous, explanation of this association is therapeutic
`failure due to insufficient plasma levels of an-
`tidepressants metabolized by CYP2D6 [49]. How-
`ever, alternative conceivable explanations rely on
`influences of CYP2D6 on the local brain metab-
`olism of the drug or endogenous substrates such
`as 5-methoxytryptamine [45]. This example high-
`lights the complementarities of research on liver-
`and brain-mediated effects of CYP2D6 genotypes.
`
`An approach to the investigation of the interplay of
`pharmacological and genetic effects on human
`brain function and behaviour is given by neuroi-
`maging techniques, where the brain correlates of
`genetic polymorphisms are assessed by functional
`tasks activating key networks [50]. Recently,
`
`174 ª 2014 The Association for the Publication of the Journal of Internal Medicine
`Journal of Internal Medicine, 2015, 277; 167–177
`
`neuroimaging approaches have detected differ-
`ences in healthy individuals in vivo, thus providing
`direct evidence of a functional effect of CYP2D6
`polymorphism in man. In a first study, an effect of
`CYP2D6 polymorphism was detected on rest per-
`fusion levels in the thalamus, hypothalamus, in
`the posterior cerebral cortex, and in isolated parts
`of the orbitofrontal cortex and the medial temporal
`lobe [51] (Fig. 2).
`
`In the second study [52], the effect of the CYP2D6
`polymorphism was detected on the signal elicited
`by a standard working memory task [53], and by a
`recognition of facial expressions task [54]. In both
`paradigms, the association with CYP2D6 genetic
`polymorphism affected visual areas in the posterior
`cerebral cortex. The fact that these two tasks have
`little in common suggests functional involvement
`with a basic brain function such as the capacity to
`sustain vigilance levels, which is required for
`prolonged performance [55]. These studies, how-
`ever, may have provided evidence on only one
`aspect of CYP2D6 activity, whose full role in local
`brain metabolism is yet to be precisely character-
`ized.
`
`Studies of brain perfusion at rest have shown
`that psychoactive substances may affect
`the
`brain according to different regional patterns.
`Antipsychotics, for example, have been shown to
`increase of metabolism and perfusion in the
`basal ganglia, and at a variable degree to
`decrease them in the cortex, especially in the
`frontal
`lobes [56, 57]. Likewise, comparative
`studies of brain perfusion at rest of antidepres-
`sants with different profiles show different perfu-
`sion patterns dependent on drug-target profile
`(Fig. 3,
`[58]). However, neuroimaging studies
`attempting to use neuroimaging quantitative
`assays of brain function to predict response and
`assist clinical decision-making have been few
`[50]. One recent study combined genotyping and
`neuroimaging techniques to identify reduced
`antidepressant response in carriers of a polymor-
`phism in the cannabinoid receptor gene [59]. As
`in studies of associations with outcome variables,
`careful study designs will be needed in the future
`to validate neuroimaging techniques as predictors
`of therapeutic interventions. Furthermore, beyond
`the use of endophenotypic biological markers,
`data on long-term parameters such as hospital-
`ization time, quality of life, disability or survival
`are warranted to finally estimate the cost–benefit
`ratio of pharmacogenetic testing.
`
`Vanda Exhibit 2042 - Page 8
`
`

`

`J. Stingl & R. Viviani
`
`Review: Pharmacogenetics of psychotropic drugs
`
`Conclusion
`
`Despite the efforts put in genomewide association
`studies based on data from large clinical trials,
`genotyping for CYP2D6 and CYP2C19 in psychi-
`atric patients is the only pharmacogenetic test
`whose use in clinical practice is justified by the
`available evidence. However,
`it allows only the
`prediction of
`individual differences
`in drug
`metabolism for the optimization of dosage to
`prevent drug toxicity. Accurate prediction of
`treatment response in complex psychiatric disor-
`ders is likely to require a suite of tools repre-
`senting intermediate phenotypes, as they may
`emerge from renewed efforts to understand the
`fundamental mechanisms from which psychiatric
`symptoms originate. Neuroimaging approaches
`are also at the centre of a research thrust to
`identify biological markers that may differentiate
`between similar manifestations of mental disor-
`ders
`[60]. Functional neuroimaging
`studies
`amongst other
`intermediate phenotypes show
`promise for the detection of endophenotype mark-
`ers but have not yet reached the stage where they
`can be applied in clinical practice.
`
`Conflict of interest statement
`
`The authors have no conflict of interest connected
`to this paper.
`
`References
`
`1 Schwabe U, Paffrath D. Arzneiverordnungs-Report 2013:
`Aktuelle Daten, Kosten, Trends und Kommentare. Heidelberg:
`Springer, 2013.
`2 Trivedi MH, Rush AJ, Wisniewski SR, et al. Evaluation of
`outcomes with citalopram for depression using measure-
`ment-based care in STAR*D: implications for clinical prac-
`tice. Am J Psychiatry 2006; 163: 28–40.
`3 Uher R, Perroud N, Ng MY, et al. Genome-wide pharmacoge-
`netics of antidepressant response in the GENDEP project. Am
`J Psychiatry 2010; 167: 555–64.
`4 Ising M, Lucae S, Binder EB, et al. A genomewide association
`study points to multiple loci that predict antidepressant drug
`treatment outcome in depression. Arch Gen Psychiatry 2009;
`66: 966–75.
`5 Rush AJ, Fava M, Wisniewski SR, et al. Sequenced treatment
`alternatives to relieve depression (STAR*D): rationale and
`design. Control Clin Trials 2004; 25: 119–42.
`6 StarD, MARS, Gendep Investigators. Common genetic varia-
`tion and antidepressant efficacy in major depressive disorder:
`a meta-analysis of three genome-wide pharmacogenetic stud-
`ies. Am J Psychiatry 2013; 170: 207–17.
`7 Kirchheiner J, Nickchen K, Bauer M et al. Pharmacogenetics
`of antidepressants and antipsychotics: the contribution of
`
`allelic variations to the phenotype of drug response. Mol
`Psychiatry 2004; 9: 442–73.
`8 Gonzalez FJ, Idle JR. Pharmacogenetic phenotyping and
`genotyping. Present status and future potential. Clin Phar-
`macokinet 1994; 26: 59–70.
`9 Swen JJ, Nijenhuis M, de Boer A, et al. Pharmacogenetics:
`from bench to byte–an update of guidelines. Clin Pharmacol
`Ther 2011; 89: 662–73.
`10 Relling MV, Klein TE. CPIC: clinical pharmacogenetics imple-
`mentation consortium of the pharmacogenomics research
`network. Clin Pharmacol Ther 2011; 89: 464–7.
`11 Stingl JC, Brockmoller J, Viviani R. Genetic variability of
`drug-metabolizing enzymes: the dual impact on psychiatric
`therapy and regulation of brain function. Mol Psychiatry
`2013; 18: 273–87.
`12 Mulder H, Heerdink ER, van Iersel EE, Wilmink FW, Egberts
`AC. Prevalence of patients using drugs metabolized by
`cytochrome P450 2D6 in different populations: a cross-sec-
`tional study. Ann Pharmacother 2007; 41: 408–13.
`13 Hicks JK, Swen JJ, Thorn CF, et al. Clinical pharmacogenet-
`ics implementation consortium guideline for CYP2D6 and
`CYP2C19 genotypes and dosing of tricyclic antidepressants.
`Clin Pharmacol Ther 2013; 93: 402–8.
`14 Caudle KE, Klein TE, Hoffman JM, et al. Incorporation of
`pharmacogenomics into routine clinical practice: the Clinical
`Pharmacogenetics Implementation Consortium (CPIC) guide-
`line development process. Curr Drug Metab 2014; 15: 209–17.
`15 Surh LC, Pacanowski MA, Haga SB, et al. Learning from
`product labels and label changes: how to build pharmacog-
`enomics into drug-development programs. Pharmacogenom-
`ics 2011; 11: 1637–47.
`16 Administration UFaD. Table of Pharmacogenomic Biomar-
`kers in Drug Labels
`17 Hiemke C, Baumann P, Bergemann N, et al. AGNP consensus
`guidelines for therapeutic drug monitoring in psychiatry
`update 2011. Psychopharmacology 2011; 44: 195–235.
`18 Bijl MJ, Visser LE, Hofman A et al. Influence of the CYP2D6*4
`polymorphism on dose, switching and discontinuation of
`antidepressants. Br J Clin Pharmacol 2008; 65: 558–64.
`19 Chou WH, Yan FX, de Leon J, et al. Extension of a pilot study:
`impact from the cytochrome P450 2D6 polymorphism on
`outcome and costs associated with severe mental illness. J
`Clin Psychopharmacol 2000; 20: 246–51.
`20 Rau T, Wohlleben G, Wuttke H et al. CYP2D6 genotype:
`impact on adverse effects and nonresponse during treatment
`with antidepressants-a pilot study. Clin Pharmacol Ther 2004;
`75: 386–93.
`21 Kwadijk-de Gijsel S, Bijl MJ, Visser LE, et al. Variation in the
`CYP2D6 gene is associated with a lower serum sodium
`concentration in patients on antidepressants. Br J Clin
`Pharmacol 2009; 68: 221–5.
`22 Ruano G, Szarek BL, Villagra D, et al. Length of psychiatric
`hospitalization is correlated with CYP2D6 functional status in
`inpatients with major depressive disorder. Biomark Med
`2013; 7: 429–39.
`23 Niznik HB, Tyndale RF, Sallee FR et al. The dopamine
`transporter
`and
`cytochrome
`P45OIID1
`(debrisoquine
`4-hydroxylase) in brain: resolution and identification of two
`distinct
`[3H]GBR-12935 binding proteins. Arch Biochem
`Biophys 1990; 276: 424–32.
`24 Nebert DW. Proposed role of drug-metabolizing enzymes:
`regulation of steady state levels of the ligands that effect
`
`ª 2014 The Association for the Publication of the Journal of Internal Medicine
`Journal of Internal Medicine, 2015, 277; 167–177
`
`175
`
`Vanda Exhibit 2042 - Page 9
`
`

`

`J. Stingl & R. Viviani
`
`Review: Pharmacogenetics of psychotropic drugs
`
`growth, homeostasis, differentiation, and neuroendocrine
`functions. Mol Endocrinol 1991; 5: 1203–14.
`25 Dutheil F, Beaune P, Loriot MA. Xenobiotic metabolizing
`enzymes in the central nervous system: contribution of
`cytochrome P450 enzymes in normal and pathological human
`brain. Biochimie 2008; 90: 426–36.
`26 Miksys S, Rao Y, Hoffmann E, Mash DC, Tyndale RF. Regional
`and cellular expression of CYP2D6 in human brain: higher
`levels in alcoholics. J Neurochem 2002; 82: 1376–87.
`27 Haining RL, Nichols-Haining M. Cytochrome P450-catalyzed
`pathways in human brain: metabolism meets pharmacology
`or old drugs with new mechanism of action? Pharmacol Ther
`2007; 113: 537–45.
`28 Nishimura M, Naito S. Tissue-specific mRNA expression
`profiles of human phase I metabolizing enzymes except for
`cytochrome P450 and phase II metabolizing enzymes. Drug
`Metab Pharmacokinet 2006; 21: 357–74.
`29 Tukey RH, Strassburg CP. Human UDP-glucuronosyltransfe-
`rases: metabolism, expression, and disease. Annu Rev Phar-
`macol Toxicol 2000; 40: 581–616.
`30 Miksys SL, Tyndale RF. Drug-metabolizing cytochrome P450s
`in the brain. J Psychiatry Neurosci 2002; 27: 406–15.
`31 Chinta SJ, Pai HV, Upadhya SC, Boyd MR, Ravindranath V.
`Constitutive expression and localization of the major drug
`metabolizing enzyme, cytochrome P4502D in human brain.
`Brain Res Mol Brain Res 2002;

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket