throbber
Review Series on Pharmacogenetics edited by Prof M. Pirmohamed
`
`Molecular genetics of CYP2D6: Clinical relevance with focus on
`
`psychotropic drugs
`
`
`
`,Mar]’a-LiisaDahl,PerDalén&AyrrranAl-Shtrrlfaji
`‘‘
`Department of Medical Laboratory Sciences 8 Technology, Division of Clinical Pharmacology, Karolinska Institutet, H1/iddinge University Hospital,
`SE-141 86 Stockholm, Sweden
`
`Cytochrome P450 CYPZD6 is the most extensively characterized polymorphic drug-
`metabolizing enzyme. A deficiency of the CYPZD6 enzyme is
`inherited as an
`autosomal recessive trait; these subiects (7% of Caucasians, about 1% of Orientals) are
`classified as poor metabolizers. Among the rest (extensive metabolizers), enzyme
`activity is highly variable, from extremely high in ultrarapid metabolizers, to markedly
`reduced in intermediate metabolizers. The CYPZD6 gene is highly polymorphic,
`with more than 70 allelic variants described so far. Of these, more than 15 encode
`an inactive or no enzyme at all. Others encode enzyme with reduced, ‘normal’ or
`increased enzyme activity. The C YPZD6 gene shows marked interethnic variability,
`with interpopulation differences in allele frequency and existence of ‘population-
`speciflc’ allelic variants,
`for instance among Orientals and Black Africans. The
`CYPZD6 enzyme catalyses the metabolism of a large number of clinically important
`drugs
`including antidepressants, neuroleptics,
`some antiarrhythmics,
`lipophilic
`(3—adrenoceptor blockers and opioids. The present—day knowledge on the influence
`ofthe genetic variability in CYPZD6 on the clinical pharmacokinetics and therapeutic
`effects/adverse effects of psychotropic drugs is reviewed.
`
`Keywords: antidepressants, CYPZD6, dehrisoquine, neuroleptics, polymorphism
`
`.
`Introduction
`
`and books related to various aspects of this are available
`.
`for further reading [4—8].
`
`Many drugs, especially lipophilic compounds such as
`psychotropics need to be metabolized prior to excretion
`in urine. Oxidative phase I catalysed metabolism by
`cytochrome P450 (CYP) enzyines plays a major role in
`this respect In the 1960s it was Shown that the 30_ to
`40-fold \
`antidepressant nortriptyline in patients treated with the
`same dose is due to a pronounced variation in the rate of
`metabolism of the drug [1, 2]. Twin studies further
`showed that the rate of metabolism had a strong genetic
`component [2] and in 1980 the l0—hydroxylation of nor-
`triptyline was shown to be catalysed by the polymorphic
`debrisoquine/sparteine hydroxylase (CYPZD6)
`This short review deals with the molecular genetics of
`CYP2D6 and its clinical relevance. Many recent reviews
`
`Correspondence: Professor Leif Bertilsson, Department of Medical Laboratory
`Sciences & Technology, Division of Clinical Pharmacology, Karolinska lnstitutet,
`Huddinge University Hospital, SE-|4| 86 Stockholm, Sweden. Tel.:+46
`8585 8|0 7|; Fax:+46 8585 8|0 70; E-mail: Leif.Berti|sson@|abtekki.se
`
`Received 23 February 200], accepted 24 October 200].
`
`The CYPZD6 polynqorphisnq
`
`The discovery of the debrisoquine/sparteine hydroxylation
`polymorphism
`
`In 1977, the hydroxylation of the antihypertensive drug
`dcbrisoquinc was shown to be polymorphic in nature
`[9, 10]. Independently, Eiclielbaum et al.
`[11] showed
`that the oxidation of sparteine was also polymorphic. The
`metabolic ratios
`(MR; parent driig/ metabolite) of the
`two drugs were closely correlated [12], showing that
`the same enzyme, now termed CYPZD6, was responsible
`for the two metabolic reactions.
`
`of
`(PM)
`of poor metabolizers
`incidence
`The
`debrisoquine/sparteine with deficient CYPZD6 activity
`has been investigated in many populations,
`in most of
`them with a fairly small number of subjects [13, 14].
`Among 1011 Swedish Caucasians we found 69 (6.3%)
`PM of debrisoquine [15]. This incidence is very similar
`to that in other European Caucasian populations (7—10%)
`
`@2002 Blackwell Science Ltd Br] Clin Pharmacol, 53,
`
`I
`
`I
`
`|—|22
`
`l"l"l
`
`ROXi|op00148033
`
`Vanda Exhibit 2036 - Page 1
`
`Vanda Exhibit 2036 - Page 1
`
`

`
`L. Bertilsson et al.
`
`In collaboration with Lou and associates in
`[13, 14].
`Beijing it was shown that the incidence of PM among
`695 Chinese was only 1.0% using the antimode established
`in Caucasian populations [15]. A si111ilar low incidence of
`
`was shifted to the right compared with Swedish EM
`(P<0.01)
`[15]. This showed that
`the mean rate of
`hydroxylation of debrisoquine was lower
`in Chinese
`EM compared with Caucasian EM. This right shift in
`
`Molecular genetics of the poor metabolizers
`
`The gene encoding the CYPZD6 enzyme is localized on
`chromosome 22 [18]. Three major mutant alleles, now
`termed CYP2D6*3, *4 and *5 [23] (Table 1), associated
`with the PM phenotype, were found early on in
`Caucasians
`[19—22].
`In
`Swedish Caucasians,
`the
`CYP2D6*4 allele occurs with a frequency of 22% and
`accounts for more than 75% of the mutant alleles in this
`
`population [23]. The CYP21)6*4 allele is almost absent
`in Chinese and this is the reason for the low incidence
`
`(1%) of PM in this population compared with 6% in
`Caucasians
`[15]. The frequency of the gene deletion
`(CYP2D6*5) on the other hand is very similar, i.e. 4—6%
`in different populations (Table l). The CYPZD6 gene
` wim
`more than 70 allelic variants described so far (http://
`www.imm.ki.se/CYPalleles/cyp2d6.htm). In addition to
`the CYPZD6 *3, *4 and *5, alleles, a large number of
`low—frequency alleles associated with the PM phenotype
`have been identified. Usually a few variants account for
`most of the mutant alleles in a population. The alleles of
`importance may, however, vary between populations (see
`below), which needs to be taken into consideration when
`applying genotyping methodology in clinical research or
`patient care.
`
`CYP2D6*10 allele [24, 25] with the SNP C188T causing
`a Pro34Ser amino acid substitution and an unstable
`
`enzyme with decreased catalytic activity [25] (Figure 1).
`The frequency of this C YP2D6*1 0 allele is similar, about
`50%,
`in Chinese, Japanese and Koreans, but extremely
`low among Caucasians (Table 1).
`Masimirembwa et al.
`[27] found a right shift of the
`MR in black Zimbabweans, similar to that found in
`Orientals. A mutated allele encoding an enzyme with
`decreased activity was subsequently identified and named
`CYP2D6* I 7. The frequency of this allele was found to be
`34% in Zimbabweans [27] (Table 1), 17% in Tanzanians
`[28], 28% in Ghanaians [29] and 9% in Ethiopians [30].
`This and many other studies demonstrate the genetic
`heterogeneity of different black populations of Africa.
`There are thus three fairly population specific alleles with
`CYP2—D6*4 in Caucasians,
`*1—0 in—Asi—ans and *17 in
`Africans. These mutations must have occurred after the
`
`separation of the respective populations from each other.
`and Orientals
`In Caucasians
`a
`close
`geno— and
`phenotype relationship has been demonstrated [23, 25,
`26]. However, in studies in Ethiopia [30], Ghana [29] and
`Tanzania [28] a lower CYP2D() activity in relation to
`genotype has been demonstrated, indicating that in Africa,
`environmental factors, e.g. infections or food constituents
`are probably of importance in addition to genetic factors.
`
`Alleles in Orientals and Afiricans encoding C YPZD6 with
`decreased activity
`
`Our early studies comparing CYP2D6 activity between
`Swedish and Chinese subjects revealed that the distribu—
`wfiono th:lVI‘Re of C inese ex ensive me a oi ers
`
`Gene duplication, multiduplication and amplification as
`causes of increased CYPZD6 activity
`
`The problems in treating PM of debrisoquine have been
`extensively discussed over the years since the discovery of
`the CYP2D6 polymorphism [14]. Much less attention has
`een given 0 a ien s a
`e o
`er ex reme, i.e. u rara i
`
`Table 1 Frequency of CYP2D6*1 or *2 alleles (causing ‘normal’ enzyme activity) and some alleles causing no or deficient CYPZD6 activity in three
`different ethnic populations.
`
`C YPZD6 alleles
`
`Functional mutation
`
`Consequence
`
`Swedish
`
`Allele frequency (%)
`Chinese
`
`Zimbabwean
`
`*1 or *2
`*3
`
`*4
`*5
`*10
`*17
`
`A2637 del
`
`G1934A
`Gene dclction
`C188T
`C1111T
`
`Frame shift
`
`Splicing defect
`No enzyme
`Unstable enzyme
`Reduced affinity
`
`69
`2
`
`22
`4
`n d
`n d
`
`43
`0
`
`0—1
`6
`51
`nd.
`
`54
`0
`
`2
`4
`6
`34
`
`nd. 2 not determined.
`
`Data are from original publications [23, 25, 27] and reviews [4, 5].
`
`l—f2
`
`©2002 Blackwell Science Ltd Br] Clin Pharmacol, 53,
`
`I
`
`I
`
`|—|22
`
`ROXi|op00148034
`
`Vanda Exhibit 2036 - Page 2
`
`Vanda Exhibit 2036 - Page 2
`
`

`
`10
`
`25
`
`20
`
`15
`
`_. O
`
`U‘!
`
`_; U10
`
`01E3
`
`25
`
`20
`
`15
`
`10
`
`Numberofsubjects
`
`wt/wt
`
`mut/wt
`
`mut/mut
`
`Total
`
`Molecular genetics of CYPZD6
`
`duplication/multiduplication of the CYP2l)6*2 gene and
`low debrisoquine MR [34].
`In Swedish Caucasians the frequency of subjects having
`duplicated/inultiduplicated genes
`is about 1—2% [34].
`
`to 3.6% in Germany [35], 7—10% in Spain [36, 37] and
`10% on Sicily in Italy [38]. The frequency is as high as
`29% in black Ethiopians [30] and 20% in Saudi Arabians
`[39]. There is
`thus
`a European—African north—south
`gradient in the incidence of CYPZD6 gene duplication.
`It has been speculated that the high incidence in Spain
`and Italy may have an ancestry in the Arabian conquest
`in the Mediterranean area [36]. Caucasian subjects with
`a CYPZD6 gene duplication have been shown to be
`ultrarapid metabolizers of debrisoquine with MR usually
`between 0.01 and 0.15 [31, 34—36].
`In the study
`of Aklillii
`et al.
`[30], black Ethiopians with multiple
`CYPZD6 genes had higher MR, usually between 0.1
`and 1. These subjects do thus not have the ultrarapid
`metabolism of debrisoquine demonstrated for Caucasians
`with multiple genes. This might be due to environmental
`flaeters in—Africa causrnga decreased
`
`Clinical relevance of the CYPZD6
`
`p0Iy1’I‘l01'pI‘liSl’I‘l
`
`0.1
`
`1
`Metabolic ratio
`
`10
`
`Figure 1 Distribution of the debrisoquine MR (parent drug/
`4—hydroxy metabolite) in three genotype groups related to the
`CYP2D6*10 allele in 152 Korean subjects.
`wt = CYPZD6*1(or*Z) and mut= CYPZD6*10. Reproduced
`with permission from Roh et al. [26].
`
`In 1993, we described a Swedish family
`metabolizers.
`with the father and his daughter and son having 12 extra
`copies of a functional CYP2D6*2 gene in the CYPZD
`locus [31]. These subjects were ultrarapid inetabolizers of
`debrisoquine with MR 0.01—0.02 (Figure 2). In another
`family, duplication of the CYP2l)6*2 gene was also
`associated with extremely high CYPZD6 activity [31].
`This was the first demonstration of an inherited duplica-
`tion/amplification of an active gene encoding a drug
`metabolizing enzyme. We also described two patients,
`who had to be treated with extremely high doses of
`antidepressants [32, 33]. One of the patients is further
`described below. The CYPZD locus of these patients was
`found to contain a duplication of the CYP2D6*2 gene. A
`population study confirmed the association between the
`
`Since the discovery of the CYPZD6 polymorphism almost
`100 drugs have been shown to be substrates for this
`enzyme. Some of these drugs are shown in Table 2. The
`clinical importance of the polymorphism depends on a
`number of factors including whether the parent com-
`pound, metabolite(s) or both are metabolized or formed by
`CYPZD6; whether the parent compound,
`the metab-
`olite(s) or both are active; the potency ofthe active species;
`and the overall contribution of the CYP2D6—dependent
`pathway to the clearance of the drug. Furthermore, the
`therapeutic index of the drug (narrow—broad), possible
`saturation of the CYP2D6—dependent pathway,
`and
`th%ee
`to be considered. Thus, the clinical impact of CYPZD6
`dependent metabolism needs to be carefully investigated
`for each substrate. So far, the majority of in viva data on
`the role of CYPZD6 are from single—dose pharmacokinetic
`The
`studies.
`increasing
`availability of genotyping
`methods has made clinical studies in patients receiving
`therapeutic doses possible. We will here highlight these
`aspects with some examples, mainly from the field of
`psychopharmacology.
`
`Nortriptyline
`
`Nortriptyline was one of the first clinically important
`drugs to be shown to be metabolized by CYPZD6 [3, 40].
`These early studies (prior to the era of genotyping) were
`
`@2002 Blackwell Science Ltd Br] Clin Pharmacol. 53.
`
`I
`
`I
`
`|—|22
`
`H3
`
`ROXi|op00148035
`
`Vanda Exhibit 2036 - Page 3
`
`Vanda Exhibit 2036 - Page 3
`
`

`
`L. Bertilsson et al.
`
`
`
`Numberofindividuals
`
`Debrisoquine/4-OH-debrisoquine
`
`10
`
`100
`
`1000
`
` USedDoses of nortriptyline
`
`500
`
`100-150
`
`‘Normal dose’
`
`10-20 mg
`
`Figure 2 Distribution of the urinary debrisoquine MR in 757 healthy Swedish subjects with schematic presentation of CYPZD6
`genotypes, where a cross in an allele indicates a detrimental mutation. Also tentative doses of nortriptyline to be used in different
`genotypes are indicated. From [64].
`
`Table 2 Some drugs whose metabolism is catalysed by CYPZD6
`
`fl—/ldrenoceptor blockers
`Metoprolol
`Propranolol
`Timolol
`
`Antiarrhythmic drugs
`Encainide
`Flecainide
`
`Perhexilene
`Propafenone
`Sparteine
`
`Antidepressants
`Amitriptyline
`Clomipramine
`Desipramine
`Fluoxetine
`
`Fluvoxamine
`Imipramine
`Mians erin
`
`Nortriptyline
`Paroxetine
`Venlafaxine
`
`Nemoleptics
`Halop eridol
`Perphenazine
`Risp ericlone
`Thioridazine
`
`Zuclopenthixol
`
`Zvliscellaneous
`
`Codeine
`Debrisoquine
`Dextromethorphan
`Phenforniin
`Tolterodine
`Tramadol
`
`performed in phenotyped panels of healthy subjects and
`the results have been confirmed in 1217120 in patients as well
`as in Vitro using human liver microsomes and expressed
`enzymes. In a recent study by Dalen et al.
`[41], nor-
`triptyline was given as a single oral dose to 21 healthy
`Swedish Caucasian subjects with different CYPZD6
`genotypes. As seen in Figure 3, there was a decrease in
`the plasma concentrations of nortriptyline from subjects
`with 0 functional genes (CYP2D6*4/*4 genotype)
`to
`those with 1, 2 and 3 (gene duplication) functional genes.
`
`The plasma concentrations of the parent drug were
`extremely low in one subject with 13 CYP2D6*2
`
`10—hydroXynortriptyline showed the opposite pattern, i.e.
`highest concentrations in the subject with 13 functional
`genes and lowest in the PM (Figure 3). This study clearly
`shows the impact of the detrimental C YP2D6*4 allele as
`well as of the duplication/amplification of the C YP2D6*2
`gene on the metabolism of nortriptyline [41]. A relation-
`ship between the CYPZD6 genotype and steady—state
`plasma concentrations of nortriptyline has
`also been
`
`demonstrated in Swedish depressed patients treated with
`nortriptyline [42].
`[41]
`We used the single dose results of Dalen et Ell.
`(Figure 3)
`to simulate steady—state concentrations of
`nortriptyline in the different genotype groups
`after
`different daily doses of the drug assuming linear kinetics
`(Figure 4). A dose of 25 mg three times daily, which is
`usually recommended as
`a starting dose,
`resulted in
`concentrations near the upper limit of the recommended
`therapeutic interval
`(200—60O nivi)
`in subjects with 0
`(PM) and 1
`(heterozygotes) functional CYPZD6 genes
`(Figure 4, upper curves). Subjects with 2 or more
`functional genes
`fall below 200 nM. At
`the usually
`recommended daily dose of nortriptyline 150 mg, subjects
`with O or 1 functional genes would attain levels above
`
`FI4
`
`©2002 Blackwell Science Ltd Br] Clin Pharmacol, 53,
`
`I
`
`I
`
`|—|22
`
`ROXi|op00148036
`
`Vanda Exhibit 2036 - Page 4
`
`Vanda Exhibit 2036 - Page 4
`
`

`
`Nortriptyline
`
`10-hydroxynortriptyline
`
`250
`
`Molecular genetics of CYP2D6
`
`Number of
`functional
`C’Y‘P2D6genes
`
`Number of
`functional
`
`CYP2D6 genes
`
`200
`
`150
`
`100
`
`50
`
`60
`
`50
`
`40
`
`30
`
`20
`
`[9
`
`E
`5
`5 <1)
`2 V’
`~55
`-8
`«E l—
`8 E)
`g E
`0 '53
`“’
`‘-
`§ 3En.
`
`
`
`
`
`24
`
`48
`
`72
`
`hydroxynortriptyline (right) in dilferent CYPZD6 genotype groups
`Figure 3 Mean plasma concentrations of nortriptyline (left) and 10-
`after a single oral dose of nortriptyline. The numerals close to the curves represent the number of fianctional CYP2D6 genes in each
`genotype group. In groups with 0-3 fianctional genes, there were fi
`ve subjects in each group while there was only one subject with
`[41]
`
`
`
`Time (h)
`
`600 nM and might therefore be at higher risk of devel-
`oping adverse drug reactions. Subjects with 2 functional
`genes, who constitute about half of Caucasian populations,
`are in the middle of the therapeutic interval (Figure 4,
`middle
`curves).
`Subjects with gene duplication or
`amplification might
`require increased doses of nor-
`triptyline,
`e.g. 75 mg three
`times daily (Figure
`4,
`bottom curves) or higher. One out of 10 patients in
`Italy and Spain, where gene duplication is common, might
`require increased doses of CYP2D6 substrates. It should be
`underscored that the curves presented in Figure 4 are
`simulated from the single dose data of Figure 3 assuming
`linear kinetics. Early studies by Alexanderson [43] showed
`that single dose data of nortriptyline can be used to predict
`
`kinetics of this drug seem to occur in extensive meta-
`bolizers when high doses of nortriptyline saturate the
`capacity for hydroxylation [44, 45].
`It should also be
`remembered that the 10—hydroxy metabolite of nortripty-
`line,
`formed by CYP2D6,
`is pharmacologically active
`although its relative contribution to the clinical effect and
`toxicity of nortriptyline has not been clearly elucidated
`[46]. The contribution of this metabolite is probably
`more important in ultrarapid metabolizers than in other
`patients.
`Using the same protocol as in the study ofDalén er al.
`[41], we investigated the influence ofthe Oriental—speciflc
`CYP2D6*10 allele on the disposition of nortriptyline in
`Chinese subjects living in Sweden [47]. Recently, Morita
`et al. [48] showed the influence of the CYP2D6*10 allele
`
`on the steady—state plasma levels of nortriptyline and its
`l0—hydroxy metabolite in Japanese depressed patients.
`studies
`From these two
`it may be concluded that
`CYP2D6*10 encodes an enzyme with decreased activity
`to metabolize nortriptyline. This effect is less pronounced
`than that of the Caucasian—specific CYP2D6*4 allele,
`which encodes no enzyme at all.
`Genotyping or phenotyping for CYP2D6 may be a
`tool
`to predict proper initial dosing of drugs such as
`nortriptyline in individual patients, especially those with
`extremely low (PM) or high
`CYP2D6 activity. This
`can be demonstrated with our experience with two
`patients for whom the dosage of nortriptyline and other
`antidepressants needed to be individualized (Figure 2)
`[37, 33, 40, R0]
`Patient 1. A ()9 year old woman was hospitalized for
`moderate to severe depression and treated with nortripty—
`line in a modest dose of 25 mg three times daily. Two days
`after the start of treatment she complained of dizziness
`[49]. After a further 6 days of treatment, she complained
`of increasing tiredness and vertigo and appeared slightly
`confused. Low clearance of nortriptyline was suspected,
`blood was
`taken for nortriptyline analysis
`and the
`dosage was decreased to 25 mg once daily. The plasma
`concentration of nortriptyline after 8 days of treatment
`with the 75 mg daily dose was 1300 nM (recommended
`plasma concentration range 200—600 nM). The concentra—
`tion on 25 mg daily for 12 days was 742 nM. When the
`dosage was further reduced to 20 mg at night, the patient
`had no side—effects and made an excellent recovery [49].
`
`@2002 Blackwell Science Ltd Br] Clin Pharmacol. 53.
`
`I
`
`I
`
`|—|22
`
`l"lT>
`
`ROXi|op00148037
`
`Vanda Exhibit 2036 - Page 5
`
`Vanda Exhibit 2036 - Page 5
`
`

`
`L. Bertilsson et al.
`
`25mg ><3
`
`1 000
`800
`600 .
`V
`
`/1'
`
`f
`N
`fhlrrl2>[t)i(?>h(el|
`CYP2D6 genes
`
`/////////I/I/II///
`
`400 //n///m/Jn/// (1)
`
`-/
`
`' ’
`
`200
`0 -
`2
`0
`50 mg x 3
`
`4
`
`6
`
`8
`
`10
`
`%2
`$84
`12
`
`is also partly metabolized by CYPZD6, it was not sur-
`prising that this patient also had low plasma concentrations
`of amitriptyline and its metabolite nortriptyline when
`treated with amitriptyline. In 1993, 8 years after we flrst
`9
`
`she had a duplication of the CYPZD6 gene and thus
`3 functional genes,
`leading to increased activity of the
`enzyme [33].
`
`Other antidepressants
`
`CYPZD6 has been shown to catalyse the metabolism ofa
`number of other antidepressants including the serotonin-
`reuptake inhibitors paroxetine, fluvoxamine and fluox—
`etine as well as venlafaxine and mianserin (Table 2). As
`an example, Sindrup et al. [51] found 25-fold differences
`in plasma concentrations of paroxetine between PM and
`EM after a single oral dose of the drug. The inter-
`phenotype difference was, however, only two—fold at
`steady—state, due to paroXetine’s saturable metabolism
`catalysed by CYPZD6 m EM.
`et a-l.
`[52] also
`found two—fold higher median steady—state plasma con-
`centrations of paroxetine in heterozygous EM than in
`homozygous EM subjects, with a considerable overlap in
`the distribution of paroxetine concentrations between the
`two genotypes. Unfortunately, no PM or UM subjects
`were included in that study. The clinical significance ofthe
`CYPZD6 genotype on the steady state plasma levels of
`fluoxetine and fluvoxamine is not known. Venlafaxine
`
`undergoes CYPZD6 dependent metabolism to the active
`major metabolite O—desmethylvenlafaXine, while
`its
`N—desmethylation is catalysed by CYP3A4, and possibly
`by CYPZC19 and CYPZC9 [53, 54]. PM ofCYP2D6 had
`a more than 4-fold lower oral clearance of venlafaxine
`
`"A
`E
`5
`g 1000
`-E
`800
`g 600
`/1
`
`2
`1/
`’/
`7 "
`4r
`4,4,
`E
`/1’
`
`E 200 /.1’, 3/4
`~
`'1;
`’
`13
`O
`g
`12
`
`>
`
`A_
`
`0
`1
`
`Q_
`
`0
`
`2
`
`4
`
`6
`
`8
`
`10
`
`§100075mg><3 01
`
`800
`
`I
`V
`
`600 V
`400 /"
`200
`O
`
`0
`
`j
`
`,.1« ,
`
`1
`
`1»
`
`" » "
`
`1*
`
`j
`
`4»
`
`” r
`
`A)
`
`4
`
`2
`
`4
`
`6
`
`8
`
`10
`
`WHEWHVS)
`
`2
`3/4
`
`13
`12
`
`Figure 4 SLeady—sLaLe levels of nortriptyline simulated from the
`single dose results of Dalen et al. [41] in different CYPZD6
`genotypes. The shaded area indicates the commonly
`recommended ‘therapeutic’ range of 200—6OO nM.
`
`compared to EM, mainly due to a decreased capacity to
`form the O—desmethylated metabolites [55]. In a study
`Thirty days later, after she had stopped taking nortripty—
`on 33 patients with depression treated with 225 mg
`hP€,Eh€p%EPtW%—ph€P9Wp@dW¢h the
`She had a high metabolic ratio of43 and was thus classified
`CYPZD6
`genotype
`and
`the
`ratio
`O—desmethyl—
`as a PM of debrisoquine.
`venlafaxine/venlafaxine was found, PM having extremely
`Patient 2. A 41 year old woman had been treated
`low ratios and UM high ratios compared with lio111o—
`for
`long periods with high doses
`of nortriptyline
`and heterozygous EM [56]. A significant
`relationship
`(300—5OO mg day_1) to achieve ‘therapeutic’ plasma levels
`between the CYPZD6 genotype (ie.
`the presence of
`(200—()00 nM)
`[32].
`The mean (is.d.) plasma level in
`the C YP2D6*10 allele) and the plasma kinetics of venla—
`seven samples drawn at a dose of 300 mg day_1 was
`faXine and O—desmethylvenlafaXine has also been shown
`in Japanese subjects
`[57]. A relationship between the
`291 i56 nM. The plasma concentration of unconjugated
`10—hydroxynortriptyline was about 10 times higher than
`PM phenotype and cardiovascular toxicity of venlafaxine
`that of the parent drug, which is much higher than
`has been suggested, based on four patients with PM
`usual. A debrisoquine phenotyping test
`confirmed
`phenotype which was either genetically determined or
`that
`this patient
`is an ultrarapid metabolizer of debri—
`due to inhibition of CYPZD6 activity by concomitant
`soquine (and nortriptyline) with a metabolic ratio of
`drugs [55]. The side—effects reported included palpita-
`0.07 [32]. This metabolic ratio was one of the lowest
`tions, shortness of breath and proarrhythmias due to the
`seen in a Swedish population (Figure 2). As amitriptyline
`heterogeneity in cardiac repolarization.
`
`FI6
`
`©2002 Blackwell Science Ltd Br] Clin Pharmacol, 53,
`
`I
`
`I
`
`|—|22
`
`ROXi|op00148038
`
`Vanda Exhibit 2036 - Page 6
`
`Vanda Exhibit 2036 - Page 6
`
`

`
`Halopei/idol
`
`[58] gave low single oral doses (2—4 mg)
`Llerena et al.
`of lialoperidol
`to panels of six EM and six PM of
`
`than EM, the mean plasma half—life being longer (29.4 and
`16.3 h, respectively; P<0.01) and the mean clearance
`lower (1.16 and 2.49 l h_1 kg_1, respectively; P< 0.05)
`[58]. The plasma level of reduced haloperidol was also
`higher in PM than in EM [59]. In a clinical study involv-
`ing eight Caucasian patients with schizophrenia treated
`with depot haloperidol
`(decanoate),
`the dopamine D2
`receptor occupancy was determined by positron emission
`tomography 1 and 4 weeks after intramuscular injection
`of the drug [60]. One of the patients was genotypically
`a PM of debrisoquine. Of the group, he had the highest
`plasma concentration of haloperidol and also the highest
`D2 receptor occupancy.
`Two studies
`from Japan [61, 62] have shown a
`relationship between increased steady state haloperidol
`plasma concentrations and the presence of CYP2D6*10
`(ane1*5)
`1°mg
`daily oral doses of haloperidol. In a recent study in Korea
`[63],
`a
`relationship between haloperidol and reduced
`haloperidol concentrations and CYPZD6 genotype was
`established in patients receiving less than 20 mg halo-
`peridol daily, but not in patients receiving higher doses.
`We believe that the high aff1nity—low capacity CYPZD6
`plays an important role at low concentrations/ doses of
`haloperidol, while the low afflnity—high capacity CYP3A4
`becomes more important at higher doses. Thus,
`the
`importance of the CYPZD6 genotype depends on the
`dose range used. The metabolic pathway of haloperidol
`catalysed by CYPZD6 is presently not known.
`
`Other neuroleptics
`
`Also many other neuroleptics
`
`such as perphenazine,
`
`shown to be metabolized by CYPZD6 (Table 2) [64]. At
`steady state,
`the median oral clearance of perphenazine
`was tliree—fold lower in PM than in homozygous EM
`of debrisoquine [65]. Similarly,
`there was a two—fold
`difference in the clearance of zuclopenthixol in the two
`genotype groups [65]. That PM reach higher average
`plasma concentrations per dose unit of perphenazine and
`zuclopenthixol than EM has been confirmed by others
`[66, 67]. All these studies have only included patients on
`oral treatment. We have recently demonstrated that the
`difference between PM and EM in steady—state plasma
`levels of zuclopenthixol is also seen during treatment with
`intramuscular zuclopenthixol decanoate [68].
`The metabolism of risperidone to its active metabolite
`9—hydroxyrisperidone is mainly dependent on CYPZD6,
`
`Molecular genetics of CYPZD6
`
`leading to significantly higher dose—corrected steady—state
`plasma levels of the parent drug in PM than in EM [38]
`(Figure 5). However,
`the concentrations of the ‘active
`moiety’, i.e. the su111 of risperidone and 9—liydroxyrisper—
`
`(Figure 5). Thus, as the parent compound and metabolite
`are considered pharmacologically equipotent,
`the poly-
`morphic metabolism is in this case not expected to be of
`clinical significance. It
`is to be pointed out that in all
`the published studies,
`there was a large overlap in the
`steady state plasma concentrations of neuroleptics between
`the different genotype groups,
`indicating that other
`factors in addition to the CYPZD6 genotype are of
`major importance for the interindividual variability in
`pharmacokinetics.
`
`T/4empezm'c elfkcts/side-elfects and C YPZD6 genotype
`
`Although the pharmacokinetic consequences of poly-
`morphic metabolism are relatively well documented for
`a number of CYPZD6 substrates, its clinical impact with
`
`and mainly based on case reports (see above). The clearest
`example of clinical significance relates to the antianginal
`drug perhexilene. Patients with perhexilene—induced
`peripheral neuropathy achieved higher blood concentra-
`tions than patients without this adverse effect [69]. In a
`retrospective study it was shown that of 20 patients
`developing peripheral neuropathy while taking this drug,
`11 were PM (55%) and ofthe 9 EM, 6 had a debrisoquine
`MR higher than 1
`[70]. The polymorphism apparently
`contributed to the withdrawal of perhexilene from the
`market in many countries in 1980s, but in patients with
`‘normal’ debrisoquine metabolism, other factors probably
`contributed to the development of the neuropathy.
`A number of studies have explored the relationship
`between adverse
`effects of psychotropic drugs
`and
`CYPZD6 genotype. For example, Chen 8: coworkers
`
`[71]féHflda 6f‘ "
`CYPZD6 alleles
`in depressed patients with adverse
`reactions compared with those without such reactions.
`The possible association between the CYPZD6 genotype
`and neuroleptic—induced movement disorders has been
`addressed in several studies [72—79] (Table 3). The results
`have been inconsistent but tend to show a slight over-
`representation of mutated C YPZD6 alleles in patients with
`tardive dyskinesia and parkinsonism during neuroleptic
`treatment. Possible explanations for the partly inconsistent
`or negative results are that in some studies the patients
`were taking various neuroleptics including those not
`metabolized by CYPZD6, or on polytherapy with drugs
`that are potent inhibitors of CYPZD6. In a recent 1 year
`follow—up pilot study of 100 consecutive psychiatric
`in—patients genotyped for CYPZD6 on
`admission,
`
`@2002 Blackwell Science Ltd Br] Clin Pharmacol. 53.
`
`I
`
`I
`
`|—|22
`
`H7
`
`ROXi|op00148039
`
`Vanda Exhibit 2036 - Page 7
`
`Vanda Exhibit 2036 - Page 7
`
`

`
`L. Bertilsson et al.
`
`25
`
`a
`
`80
`
`b
`
`A
`
`‘:37
`E
`L
`6
`E
`E
`@
`g
`E
`-§
`D:
`
`20
`
`15
`
`10
`

`
`8
`
`9
`
`o
`
`80
`§o
`
`0
`
`3
`
`5
`
`O
`
`8
`o
`8
`:2
`
`oooogoo
`8
`80
`EM
`EM
`UM
`PM
`homozygous heterozygous
`
`:‘~ 70
`
`E”
`60
`_
`l:
`50
`2
`5
`Q 40
`L;
`E;
`E
`20
`%:
`< 10
`O
`
`30
`

`

`O
`
`o
`
`O
`O
`
`O
`
`0
`028
`000

`0
`O
`
`EM
`UM
`EM
`PM
`homozygous heterozygous
`
`0
`0
`
`o
`
`o
`0
`o
`880
`0
`80
`
`00
`
`Figure 5 Relationship between the CYPZD6 genotype and the plasma concentration—to—dose (C/D) ratios of risperidone (left) and the
`active moiety of risperidone (risperidone plus 9—hydroxyrisperidone, right). UM=ultrarapid metabolizers, defined as a carrier of gene
`duplication; EM homozygous=extensive metabolizers homozygous for the functional CYP2D6*1 allele; EM heterozygous=EM
`heterozygous for a defective CYPZD6 gene; PM=poor metabolizers carrying two defect genes. Reproduced with permission fi:om
`Scordo at al. [38].
`
`Table 3 Studies on the relationship between CYPZD6 genotype
`and neuroleptic—induced movement disorders.
`
`Study
`
`Side—efi’ect
`
`[72]
`Arthur et alt
`Arnistrong at alt [73]
`
`Andreasson et alt [74]
`
`Kapitany et alt [75]
`Ohmori et alt [76]
`Vandel et alt [77]
`
`[ 8]
`Flameiin et alt
`Scordo et alt
`[79]
`
`Tardive dyskinesia
`Acute dystonia
`Q]-_-H;9¥H’ mox m ne-
`
`disorders
`Tardive dyskinesia
`
`Parkinsonism
`Akathisia
`
`Tardive dyskinesia
`Tardive dyskinesia
`Extrapyramidal side—
`effects
`
`Not defined
`Extrapyramidal side—
`effects
`
`Antiarrhythmic drugs have a narrow therapeutic index
`and many of them (flecainide, encainide, propafenone,
`mexiletine, N—propyl—ajmaline) are CYPZD6 substrates.
`PM might thus be expected to have an increased risk of
`concentration—dependent adverse effects. Encainide is an
`exception as
`its O—desmetyl metabolite,
`the formation
`of which is catalysed by CYPZD6,
`is 6—1O ti111es 111ore
`N9%%
`tendency)
`[81]. Although PM treated with encainide develop less
`No (non—sign
`QRS and QT prolongation than EM, both phenotype
`tendency)
`groups
`showed a
`similar antiarrhythmic response at
`comparable doses [82]. An increased frequency of fatal
`nervous system adverse effects has been reported in PM
`during propafenone treatment
`[83].
`It has also been
`suggested that determination of
`the phenotype
`(or
`genotype) of patients before therapy with N—propyl—
`ajmaline might be useful to select the individual optimum
`dose of the drug [84]. No prospective studies have,
`however, tested this hypothesis.
`
`Relationship with
`CYPZD6 genotype
`
`No
`No
`
`No
`No
`
`Yes
`No
`Yes
`
`No
`No
`
`[80] found a trend towards greater number
`Chou et al.
`of adverse drug effects from drugs primarily metabolized
`by CYPZD6, when one moves from UM to PM. The
`costs of treating the patients with extremes of CYPZD6
`activity (i.e. PM and UM) was on average 4000—6000
`dollars greater per year than the costs of treating patients
`with the EM or intermediate metabolizer (heterozygous
`EM) genotypes. The total duration of hospital stay was
`also longer for patients in the PM group. This pilot study
`needs a large clinical
`trial to confirm these preliminary
`and interesting issues.
`
`Future perspectives
`
`The pronounced interindividual variation in the rate of
`drug metabolism has been known for many years. It was
`initially only of academic
`interest, but
`today the
`pharmaceutical industry has to document the metabolism
`of a new drug in development before registration. The
`knowledge of how a drug is metabolized and which
`enzymes are involved helps to predict drug—drug inter-
`actions and how fast an individual patient may metab-
`olize a specific drug.
`
`TF8
`
`©2002 Blackwell Science Ltd Br] Clin Pharmacol, 53,
`
`I
`
`I
`
`|—|22
`
`ROXi|op00148040
`
`Vanda Exhibit 2036 - Page 8
`
`Vanda Exhibit 2036 - Page 8
`
`

`
`Molecular genetics of CYPZD6
`
`laboratory have been
`The studies performed in the authors’
`supported by the Swedish Medical Research Council
`(3902),
`National
`Institutes of Health, USA (GM 60548-01A2)
`and
`Karolinska Institutet.
`
`or therapeutic failure. Further studies are required to
`With the high—throughput screening techniques avail-
`establis

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