SlideShare une entreprise Scribd logo
1  sur  52
linical pharmacogenomics consists of the appli-
cation of research that links measurable genetic variants
with the prediction of drug response.1 Every medical
specialty can utilize the results of pharmacogenomic
probe studies to inform the adoption of individualized
pharmacotherapy. However, psychiatric pharmacother-
apy is particularly likely to benefit from the introduction
of pharmacogenomic testing, because there are many
psychotropic agents available for selection that target
specific symptoms.
The terms pharmacogenetics and pharmacogenomics
are currently used interchangeably. However, with the
growing understanding that multiple intragenic varia-
tions should be considered in making predictions related
to medication response, the use of the term pharma-
cogenomics has become more frequently chosen to des-
ignate the process of using documented genetic variation
to guide medication selection and dosing.
Historically, psychiatrists have used empirical strategies
to select medications. In the best practices, the choice of
medications has evolved based on a rational trial-and-
error process that has used clinical indicators to select
medications and then relied on documenting treatment
responses to titrate the optimal dose for a particular
patient. Psychiatrists learn to “start low and go slow” in
order to minimize side effects. They also know that it is
6 9
P h a r m a c o l o g i c a l a s p e c t s
C
Copyright © 2010 LLS SAS. All rights reserved
www.dialogues-cns.org
Psychiatric pharmacogenomic testing in
clinical practice
David A. Mrazek, MD, FRCPsych
Keywords: pharmacogenomic testing; cytochrome P450 gene;
serotonin trans-
porter gene (SLC6A4); serotonin receptor 2A gene (HTR2A);
serotonin receptor 2C
gene (HTR2C); poor metabolizer phenotype; ultrarapid
metabolizer phenotype
Author affiliations: Chair, Department of Psychiatry and
Psychology, Mayo
Clinic; Professor of Psychiatry and Pediatrics, Mayo Clinic
College of
Medicine, Rochester, Minnesota, USA
Address for correspondence: David A. Mrazek, Mayo Clinic
College of Medicine,
200 First Street SW, Rochester, MN 55905, USA
(e-mail: [email protected])
The clinical adoption of psychiatric pharmacogenomic
testing has taken place rapidly over the past 7 years.
Initially, drug-metabolizing enzyme genes, such as the
cytochrome P450 2D6 gene (CYP2D6), were identified.
Genotyping the highly variable cytochrome P450 2D6
gene now provides clinicians with the opportunity to iden-
tify both poor metabolizers and ultrarapid metabolizers
of 2D6 substrate medications. Subsequently, genes influ-
encing the pharmacodynamic response of medications
have been made available for clinical practice. Among the
earliest “target genes” was the serotonin transporter
gene (SLC6A4) which has variants that have been shown
to influence the clinical response of patients of European
ancestry when they are treated with selective serotonin
reuptake inhibitors. Genotyping of some of the serotonin
receptor genes is also available to guide clinical practice.
The quantification of the clinical utility of pharmacoge-
nomic testing is evolving, and ethical considerations for
testing have been established. Given the increasingly clear
cost-effectiveness of genotyping, it has recently been pre-
dicted that pharmacogenomic testing will routinely be
ordered to guide the selection and dosing of psychotropic
medications.
© 2010, LLS SAS Dialogues Clin Neurosci. 2010;12:69-76.
DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 69
necessary to provide their patients with an “adequate”
trial of each medication. Unfortunately, these strategies
can result in a 3- to 4-week interval during which the
patient continues to experience symptoms. In recent
years, the potential iatrogenic harm associated with psy-
chotropic medications has become increasingly obvious,
with “black-box warnings” being attached to antide-
pressants, antipsychotic medications, stimulants, and
mood stabilizers.
Despite a growing awareness of this potential harm,
there are powerful pressures to try to accelerate the
achievement of therapeutic benefit. At the most basic
level, patients are impatient. They do not want to wait a
month to achieve symptom relief. Additionally, with an
increasing focus on the relief of specific symptoms,
strategies using multiple psychotropic medications have
become a standard of practice. Research supports the
common practice of augmenting an initial medication
with a second psychotropic drug.2 However, there is no
scientifically available evidence to support the practice
of using four or five psychotropic medications simulta-
neously. Nevertheless, patients routinely receive multi-
ple psychotropic medications in an attempt to identify
the “right combination.” While some patients do achieve
a good therapeutic response using this trial-and-error
approach to individualized medicine, it is also true that
others become overmedicated or suffer from iatrogenic
side effects.
Pharmacogenomic testing provides an innovative strat-
egy to improve the likelihood of selecting an effective
psychotropic medication. The earliest medical texts rec-
ognize that individual patients experience quite dra-
matically different responses to the same drug. There is
also a longstanding observation that unusual drug
responses can occur in members of the same family. The
identification of specific gene variants associated with
idiosyncratic responses is about 50 years old,3 and the
recognition that some psychiatric patients metabolize
antidepressants at dramatically different rates has been
documented for several decades.4 However, with the use
of newer antidepressant medications that rarely have
life-threatening complications, the relatively expensive
practice of monitoring the serum levels of newer anti-
depressant medications has become uncommon in the
United States. This change has occurred despite the fact
that serum levels of these newer agents also have dra-
matic variations based on the metabolic capacity of each
patient.
A decade ago, the cost of genotyping began to become
more affordable, and individual laboratories initiated
pharmacogenomic testing that would provide genotyping
of individual cytochrome P450 genes. However, there was
no standard or well-validated methodology for the geno-
typing of these informative genes. There was also consid-
erable variability in the interpretation of the results. In
2004, the US Food and Drug Administration (FDA)
approved the use of a new product, the AmpliChip.5 The
introduction of the AmpliChip provided reference labo-
ratories with a standard method for identifying variations
in two of the cytochrome P450 genes: cytochrome P450
2D6 (CYP2D6) and cytochrome P450 2C19 (CYP2C19).
The approval of the AmpliChip was an important land-
mark in the history of psychiatric pharmacogenomic test-
ing, and within 3 years, CYP2D6 and CYP2C19 were
being genotyped by every reference laboratory in the
country. However, this advance also highlighted some of
the challenges associated with the introduction of clinical
testing. One of the most obvious challenges that must be
addressed is how to begin to assess new variants of these
two genes in updated versions of the assay. Ideally, the
methodology for establishing drug-metabolizing pheno-
types should be updated regularly based on new molecu-
lar genetic findings showing how new genotypic variants
influence gene function. Also, the clarification of the pre-
dictive capacity of previously identified gene variants
influencing gene function is similarly evolving, and newly
identified associations between gene structure and func-
tion should ideally be incorporated into algorithms that
define the metabolic capacity of psychiatric patients.
The evolution of pharmacogenomic research should
inform modifications in pharmacogenomic testing.
However, an implication of the rapid increase in our
knowledge base is that these new studies demonstrate
limitations in the accuracy of older genotyping method-
ologies that were designed prior to the discovery of
more recent variants. What is often not well appreciated
is that even older pharmacogenomic methods provided
important information for many patients, as these early
innovations were a major advance over psychopharma-
cological practice without pharmacogenomic insights.
However, as newer methodologies have further
improved the accuracy of the prediction of medication
response, the clinical utility of pharmacogenomic testing
continues to increase.
Pharmacogenomic testing in psychiatric practice initially
focused on identifying pharmacokinetic variability that
P h a r m a c o l o g i c a l a s p e c t s
7 0
DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 70
would influence the responses of patients who had atyp-
ical genotypes. Pharmacokinetic variation influences
the concentration of a drug at its sites of action.
Pharmacogenomic testing of drug-metabolizing enzyme
genes provides a prediction of how an individual patient
will metabolize a specific psychotropic medication. More
recently, the focus of pharmacogenomic testing has
expanded to include determining variability in the phar-
macodynamic response of a patient to a specific med-
ication. This variability reflects the capacity of the indi-
vidual patient to respond to adequate exposure to the
drug. Prediction of response is estimated based on the
documentation of variations in “target genes” that code
for receptors and transporters that influence the
response of the patient to a particular medication.
This review will first identify the most widely genotyped
drug-metabolizing enzyme genes that influence the
pharmacokinetic metabolic capacity of a patient. Then,
it will focus on genes that influence the pharmacody-
namic responses of individual patients, before conclud-
ing with a brief discussion of the clinical utility of phar-
macogenomic testing and some of the ethical
considerations related to its routine use.
Pharmacogenomic testing to establish the
metabolic capacity of psychiatric patients
Many genes code for enzymes that influence drug
response. However, only the clinical implications of geno-
typing four of the most commonly tested cytochrome
P450 genes will be reviewed. The focus of this discussion
will be the clinical benefit for the patient of identifying
individualized molecular variations, and the implications
for those patients who have a quite significant decrement
in their capacity to metabolize specific psychotropic med-
ications. Identifying these individual patients provides
clinicians with a clear method of minimizing side effects.
This determination of decreased metabolic capacity is the
most obvious benefit of pharmacogenomic testing, but
implications of the pharmacogenomic testing for patients
with increased metabolic capacity will also be discussed,
as these patients are less likely to respond to specific psy-
chotropic medications.
The cytochrome P450 2D6 gene (CYP2D6)
CYP2D6 was the first drug-metabolizing enzyme gene
that was genotyped to identify psychiatric patients with
increased or decreased metabolic capacity. It is located
on chromosome 22 and consists of 4382 nucleotides.
CYP2D6 codes for an enzyme that is composed of 497
amino acids.
The CYP2D6 enzyme plays a primary role in the metab-
olism of more than 70 substrate medications, including
twelve psychotropic medications. CYP2D6 is one of the
most highly variable drug-metabolizing enzyme genes.
However, many of the other 29 P450 drug-metabolizing
enzyme genes are also highly variable. The specific
genetic variations that define variable phenotypes can
be located on a Web site maintained by the Karolinska
Institute (http://www.cypalleles.ki.se/). Each newly iden-
tified variant is included on the Web site after confir-
mation that it is unique.
There are currently 75 distinct CYP2D6 alleles posted
on this site, as well as an additional 55 CYP2D6 variants
that closely resemble one of the primary variants.
Traditionally, these variants have been classified as being
normal, deficient, or inactive drug-metabolizing alleles.
Additionally, some alleles have more recently been
demonstrated to code for an increased amount of
enzyme which enhances the metabolic activity of the
patient. Furthermore, patients can have a variable num-
ber of copies of CYP2D6. The most common number of
copies of CYP2D6 that patients carry is two. However,
some patients have only one copy and, rarely, none at all.
It is also possible to have more than two copies, and one
patient has been reported to have 13 copies.4 The devel-
opment of several different classification systems to cat-
egorize 2D6 substrate metabolic capacity of patients into
four phenotypic categories has been problematic. The
use of alternative methodologies by different research
teams has made it more difficult to study the implica-
tions of this variability.
The most important CYP2D6 phenotype to identify is
the poor 2D6 substrate metabolizer phenotype. Patients
who are poor metabolizers are at increased risk for
adverse events when they are prescribed 2D6 substrate
medications, because of their low metabolic capacity.
Patients are now classified as poor metabolizers if they
have two inactive alleles, or one inactive allele and one
deficient allele.
The second most clinically important CYP2D6 pheno-
type is the ultrarapid metabolizer phenotype. Patients
are ultrarapid metabolizers if they have either three or
more active copies of CYP2D6 or two or more enhanced
copies of CYP2D6. They are unlikely to respond to 2D6
Psychiatric pharmacogenomic testing - Mrazek Dialogues in
Clinical Neuroscience - Vol 12 . No. 1 . 2010
7 1
DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 71
substrate medications at standard doses because their
ability to rapidly metabolize these medications makes it
difficult to sustain therapeutic serum levels.
The third clinically important CYP2D6 phenotype is the
intermediate metabolizer phenotype. These patients
have one normal copy of CYP2D6, and one copy that is
either deficient or inactive. While these patients can nor-
mally benefit from 2D6 substrate medications at low-to-
moderate doses, they are at increased risk for the devel-
opment of side effects at higher doses because of their
decreased metabolic capacity, and they are more at risk
for enzyme inhibition as a consequence of drug-drug
interactions. When intermediate metabolizers are
exposed to powerful 2D6 inhibitors such as paroxetine
or fluoxetine, their metabolic capacity can be further
decreased to the level of a poor metabolizer.6
There are many psychotropic medications metabolized
by the 2D6 enzyme. Specifically, this enzyme:
• primarily metabolizes five antidepressants: fluoxetine,
paroxetine, venlafaxine, desipramine, and nortriptyline
• substantially metabolizes amitriptyline, imipramine,
doxepin, duloxetine, trazodone, and mirtazapine
• primarily metabolizes risperidone and four of the typ-
ical antipsychotic medications: chlorpromazine, thior-
idazine, perphenazine, and haloperidol
• has substantial involvement in the metabolism of arip-
iprazole and olanzapine
• primarily metabolizes atomoxetine and dextroam-
phetamine.
Beyond the prescription of psychotropic medications,
psychiatric patients are given many other 2D6 substrate
medications. Specifically, dextromethorphan is a cough
suppressant that is metabolized by the 2D6 enzyme.
Patients who are poor metabolizers of 2D6 substrate
medications are at increased risk for cognitive side effects
if taking standard doses of preparations that contain dex-
tromethorphan. Another example is codeine, which is a
prodrug. A prodrug must be converted to an active
metabolite in order to have a therapeutic effect. Patients
who are poor 2D6 metabolizers do not receive analgesic
benefit from codeine because they do not metabolize
codeine to morphine. Tamoxifen is also a prodrug that is
the most frequently prescribed treatment for breast can-
cer. Poor metabolizers have little or no benefit from
tamoxifen because they are not able to metabolize
tamoxifen to endoxifen.7,8 Additionally, paroxetine, flu-
oxetine, or bupropion should not be given to patients
who are receiving tamoxifen because they inhibit the
2D6 enzyme. Giving these inhibitors to intermediate
metabolizers can convert them to functional poor metab-
olizers. Consequently, they become unable to produce
endoxifen.9
The cytochrome P450 2C19 gene (CYP2C19)
CYP2C19 was the second drug-metabolizing enzyme
gene that was widely genotyped to identify patients with
increased or decreased metabolic capacity. It is a large
gene located on chromosome 10. It consists of 90 209
nucleotides, but codes for an enzyme that contains only
490 amino acids.
The identification of patients with low 2C19 metabolic
capacity is clinically important because it allows clini-
cians to decrease the risk of iatrogenic side effects.
The 2C19 enzyme:
• primarily metabolizes citalopram, escitalopram,
clomipramine, and amitriptyline
• has substantial involvement in the metabolism of ser-
traline, imipramine, nortriptyline, and doxepin
• plays an important role in the metabolism of clozapine
and a minimal role in the metabolism of thioridazine
• is the primary enzyme involved in the metabolism of
diazepam.
Recently, a new variant of CYP2C19 has been identified
which has enhanced function.10 Patients who are
homozygous for this new allele are less likely to respond
to 2C19 substrate medications at standard doses. The
identification of ultrarapid 2C19 metabolizers can be
helpful in evaluating patients who do not respond to
standard doses of any of these psychotropic medications.
The cytochrome P450 2C9 gene (CYP2C9)
CYP2C9 is located on chromosome 10 in relative close
proximity to CYP2C19. However, it is only about half
the size of CYP2C9 as it consists of 50 708 nucleotides.
Like CYP2C19, CYP2C9 codes for an enzyme that con-
tains 490 amino acids.
CYP2C9 is a drug-metabolizing enzyme gene that is less
routinely genotyped to identify the increased or
decreased metabolic capacity of psychiatric patients for
2C9 substrate medications. It does not play a primary
role in the metabolism of any currently prescribed psy-
chotropic medications. However, the 2C9 enzyme pro-
vides the only secondary pathway for the metabolism of
fluoxetine, so patients who are poor metabolizers of
P h a r m a c o l o g i c a l a s p e c t s
7 2
DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 72
Psychiatric pharmacogenomic testing - Mrazek Dialogues in
Clinical Neuroscience - Vol 12 . No. 1 . 2010
7 3
both 2D6 substrates and 2C9 substrates are at very high
risk for adverse effects if treated with standard dose of
fluoxetine.
The cytochrome P450 1A2 gene (CYP1A2)
CYP1A2 is a less well-studied drug-metabolizing
enzyme gene, but it codes for an enzyme that plays an
important role in the metabolism of fluvoxamine. It is
also involved in the metabolism of duloxetine and olan-
zapine. CYP1A2 is located on chromosome 15 and con-
sists of 7758 nucleotides. CYP1A2 codes for an enzyme
that is composed of 516 amino acids.
A somewhat atypical aspect of the CYP1A2 gene is that
there are alleles of this gene that are inducible by smok-
ing tobacco or consuming cruciferous vegetables, such
as cabbage or Brussels sprouts. Consequently, patients
who smoke tobacco and have two alleles of CYP1A2
that are inducible by their smoking can be difficult to
maintain on 1A2 substrate medications. A relatively
common problem occurs when these patients are treated
with olanzapine or clozapine on an inpatient psychiatric
unit that does not allow them to smoke. When they
begin to smoke after they are discharged, their serum
level drops and their psychotic symptoms often reoccur.
In some populations of European ancestry, as many as
25% of the population can have an inducible ultrarapid
CYP1A2 phenotype.
Pharmacogenomic testing to identify
variability in pharmacodynamic responses
A goal of individualized molecular psychopharmacology
is to identify medications for an individual patient that
will not only be safe, but will be effective. Progress in
making predictions of medication response has occurred,
and while the goal of being able to predict this response
with certainty has not been achieved, we can make
increasingly accurate probabilistic predictions of the
likelihood of response. Psychiatrists are familiar with this
limitation. While hundreds of randomized clinical trials
of psychotropic medications have been conducted to
identify effective psychotropic drugs, the results of these
trials only provide assurance that for a sample of
patients there is reasonable likelihood that the medica-
tion will be of more benefit than a placebo. While selec-
tive serotonin reuptake blockers are among the most
widely prescribed medications in the world, many
patients do not respond. Specifically, the largest clinical
effectiveness study of citalopram reported that less than
30% of the entire sample of patients experienced a com-
plete remission of their symptoms.11 While the ultimate
goal of pharmacodynamically designed pharmacoge-
nomic testing is to identify a drug for a specific patient
that will definitely be effective, at the current stage of
our understanding, it is only possible to identify a med-
ication that is more likely to be effective.
The serotonin transporter gene (SLC6A4)
SLC6A4 is located on chromosome 17 and consists of 37
800 nucleotides. It codes for an enzyme that is composed
of 630 amino acids.
SLC6A4 is the most widely genotyped pharmacoge-
nomic “target” gene. A meta-analysis of studies of the
relationship between the more active long form of the
indel promoter variant of this gene and responses to
selective serotonin reuptake inhibitors12 confirmed the
early finding that the long form is associated with a more
rapid and better response.13 However, this has not con-
sistently been demonstrated in patients of Asian ances-
try.14,15 The importance of ancestral heritage has been fur-
ther demonstrated by multiple analyses of the large
STAR*D effective treatment study.
Analyses that did not consider ancestral background did
not demonstrate a significant association,16 while those
that focused on patients who identified themselves as
“white” but not “Hispanic” did confirm the relationship
that patients who were homozygous for the more active
long form of the indel promoter polymorphism were
more likely to respond to citalopram. Other variants,
such as rs2553117 and the second intronic VNTR18 are
likely to influence the activity level of the gene and, con-
sequently, its response to medications that block its abil-
ity to reuptake serotonin in the synapses of the central
nervous system.
The serotonin receptor 2A gene (HTR2A)
HTR2A is located on chromosome 13 and consists of 62
663 nucleotides. Despite its large size, it codes for an
enzyme that is composed of only 471 amino acids.
There have been a series of studies examining the asso-
ciation between variants of HTR2A and antidepressant
response. A large study examining the response of
depressed patients of European ancestry to citalopram
DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 73
found that a positive response to citalopram was associ-
ated with having a copy of the adenine allele of
rs7997012.19
Another study examining a different HTR2A variant,
rs6313, reported that patients who were homozygous for
the cytosine allele were less likely to tolerate taking
paroxetine than those who had one or more copies of
the thymine allele.20
A series of studies have reported a better response to
clozapine in patients who had the thymine allele of
rs6313. The thymine allele of rs6313 has also been asso-
ciated with a lower risk for the development of
extrapyramidal side effects when taking antipsychotic
medications.21-23
The serotonin receptor 2C gene (HTR2C)
HTR2C is a very large gene that is located on the X
chromosome and consists of 326 074 nucleotides.
However, it codes for a protein product that is composed
of only 458 amino acids.
Variations in the HTR2C gene have been associated
with a better clinical response to clozapine. Specifically,
patients with schizophrenia who have a copy of the cyto-
sine allele of rs6318 have achieved better control of their
psychotic symptoms than patients with the guanine
allele.24,25 However, this same variant has been associated
with a higher risk for the development of extrapyrami-
dal side effects in patients who are taking typical antipsy-
chotic medications.26
An increased risk for the development of weight gain
has been linked to a different HTR2C variant.
Specifically, the cytosine allele of rs518147 is associated
with increased weight gain, while the thymine allele is
conceptualized as providing protection against weight
gain.27-29
The clinical utility of pharmacogenomic
testing in psychiatric practice
Assessing the clinical utility of pharmacogenomic test-
ing is an ongoing process, given that the accuracy of
genotyping is continually improving, and new research
is identifying additional genetic variants that influence
medication responses. Reports of adverse responses to
2D6 substrate medications in patients with decreased
2D6 metabolic capacity support the use of testing at this
most basic level. Specifically, poor 2D6 metabolizers
have had quite dramatic side effects to 2D6 substrate
medications3 and some toxic reactions have been
lethal.30,31 However, there have been no large random-
ized clinical trials to demonstrate the clinical utility of
pharmacogenomic testing. Such trials would reinforce
the use of testing. However, it is unlikely that these tri-
als will ever be conducted because, by definition, they
are not designed to concentrate on those patients who
are the most likely to benefit from pharmacogenomic
testing. Trials that screen vulnerable populations and
identify patients at risk for suboptimal responses to med-
ications are a more efficient method to address the clin-
ical usefulness of testing patients with decreased meta-
bolic capacity. These screened patients could then be
enrolled in protocols designed to provide optimal
response for their specific genotypes and predicted phar-
macogenomic phenotypes.
Ethical considerations for pharmacogenomic
testing in psychiatric practice
The provision of pharmacogenomic testing involves rel-
atively few risks, but ethical safeguards are still impor-
tant to consider. These are essentially the same consid-
erations that are important to think through when
ordering any laboratory test that has the potential to
direct a treatment decision.
First, clinical pharmacogenomic testing requires obtain-
ing appropriate consent. This has become a guiding prin-
ciple for all diagnostic and therapeutic procedures.
Clinicians should provide the basic rationale for pro-
ceeding with pharmacogenomic testing so that their
patients have the opportunity to provide explicit
informed consent.
Secondly, as a component of obtaining clinical consent,
it must be clear that clinical testing is a voluntary proce-
dure. This is true for virtually all clinical laboratory test-
ing with the relatively rare exceptions of mandatory test-
ing that can identify a condition with a potential
negative influence on the public health of the commu-
nity. A common example of compulsory testing is the
monitoring of infections in order to prevent contagion.
A third principle is that clinicians must insure the confi-
dentiality of sensitive medical information that becomes
a part of the medical record of the patient. This is true
whether the information is derived from a pathological
specimen that reveals a malignant carcinoma or from
magnetic resonance imaging that demonstrates atrophy
P h a r m a c o l o g i c a l a s p e c t s
7 4
DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 74
of the hippocampus. The security of the medical record
is the responsibility of the clinician.
Finally, any diagnostic medical procedure must have an
acceptable level of reliability. The degree of accuracy of
any clinical laboratory testing is dependent on a number
of key variables. Two of these variables are the serious-
ness of the prognosis for the patient if the test is positive
and the efficacy of available treatments. In designing the
treatment plan for a potentially lethal condition that is
likely to respond well to a relatively benign intervention
if it is administered early in the course of the illness, a
laboratory test with high sensitivity is desirable. The
most important objective in this situation is to identify
as quickly as possible those patients who will benefit
from treatment.
Future developments that will influence
pharmacogenomic testing in
psychiatric practice
In the 2009 presidential lecture of the American
Psychiatric Association, it was predicted that pharma-
cogenomic testing would become a part of everyday psy-
chiatric practice.32 Ironically, in many academic health
centers, pharmacogenomic testing has been utilized since
2004—the time of the introduction of the AmpliChip.
Over the intervening years, early adopters have inte-
grated pharmacogenomic testing into their inpatient
protocols and ultimately into their outpatient practices.
However, this testing has not yet been included in many
clinical guidelines.
Pharmacogenomic testing is an innovation, and it takes
time for innovations to become integrated into standard
practice. While it is difficult to predict with accuracy just
how quickly pharmacogenomic testing will become an
essential component of clinical psychopharmacological
practice, there is no question that this will happen.
Ironically, given advances in our ability to sequence genes
both rapidly and inexpensively, there will come a time in
the near future when most patients will know their 2D6
phenotype in the same way as today they know their
blood type. However, well before we reach a state of uni-
versal awareness of our informative genotypes, our
patients will no longer accept avoidable side effects, and
will demand basic pharmacogenomic testing prior to tak-
ing antidepressant or antipsychotic medications. ❏
Psychiatric pharmacogenomic testing - Mrazek Dialogues in
Clinical Neuroscience - Vol 12 . No. 1 . 2010
7 5
REFERENCES
1. Mrazek DA. Psychiatric Pharmacogenomics. New York, NY:
Oxford
University Press; 2010.
2. Thase ME, Friedman ES, Biggs MM, et al. Cognitive therapy
versus med-
ication in augmentation and switch strategies as second-step
treatments: a
STAR*D report. Am J Psychiatry. 2007;164:739-752.
3. Weinshilboum R. Inheritance and drug response. N Engl J
Med.
2003;348:529-537.
4. Dalen P, Dahl ML, Ruiz ML, Nordin J, Bertilsson L. 10-
Hydroxylation of
nortriptyline in white persons with 0, 1, 2, 3, and 13 functional
CYP2D6
genes. Clin Pharmacol Ther. 1998;63:444-452.
5. Mrazek D. Out of the pipeline: pharmacogenomic DNA chip.
Curr
Psychiatr. 2005;4:67-73.
6. Preskorn S. Reproducibility of the in vivo effect of the
selective sero-
tonin reuptake inhibitors on the in vivo function of cytochrome
P450 2D6:
an update (part II). J Psychiatr Pract. 2003;9:150-158.
7. Goetz MP, Knox SK, Suman VJ, et al. The impact of
cytochrome P450
2D6 metabolism in women receiving adjuvant tamoxifen. Breast
Cancer Res
Treat. 2007;101:113-21.
8. Schroth W, Goetz MP, Hamann U, et al. Associaiton between
CYP2D6
polymorphisms and outcomes among women with early stage
breast can-
cer treated with tamoxifen. JAMA. 2009;302:1429-1436.
9. Henry NL, Stearns V, Flockhart DA, Hayes DF, Riba M.
Drug interactions
and pharmacogenomics in the treatment of breast cancer and
depression.
Am J Psychiatry. 2008;165:1251-1255.
10. Sim SC, Risinger C, Dahl ML, et al. A common novel
CYP2C19 gene vari-
ant causes ultrarapid drug metabolism relevant for the drug
response to
proton pump inhibitors and antidepressants. Clin Pharmacol
Ther.
2006;79:103-113.
11. Trivedi M, Rush A, Wisniewski S, et al. Evaluation of
outcomes with
citalopram for depression using measurement-based care in
STAR*D: impli-
cations for clinical practice. Am J Psychiatry. 2006;163:28.
12. Serretti A, Kato M, De Ronchi D, Kinoshita T. Meta-
analysis of serotonin
transporter gene promoter polymorphism (5-HTTLPR)
association with selec-
tive serotonin reuptake inhibitor efficacy in depressed patients.
Mol
Psychiatry. 2007;12:247-257.
13. Serretti A, Zanardi R, Rossini D, Cusin C, Lilli R, Smeraldi
E. Influence of
tryptophan hydroxylase and serotonin transporter genes on
fluvoxamine
antidepressant activity. Mol Psychiatry. 2001;6:586-592.
14. Yoshida K, Itoa K, Satoa K, et al. Influence of the serotonin
transporter
gene-linked polymorphic region on the antidepressant response
to fluvox-
amine in Japanese depressed patients. Prog
Neuropsychopharmacol Biol
Psychiatry. 2002;26:383-386.
15. Kim DK, Lim SW, Lee S, et al. Serotonin transporter gene
polymorphism
and antidepressant response. Neuroreport. 2000;11:215-219.
16. Peters E, Slager S, McGrath P, Knowles J, Hamilton S.
Investigation of
serotonin-related genes in antidepressant response. Mol
Psychiatry.
2004;9:879-889.
17. Hu XZ, Rush AJ, Charney D, et al. Association between a
functional
serotonin transporter promoter polymorphism and citalopram
treatment
in adult outpatients with major depression. Arch Gen
Psychiatry. 2007;64:783-
792.
18. Mrazek DA, Rush AJ, Biernacka JM, et al. SLC6A4
variation and citalo-
pram response. Am J Med Genet Part B. 2009;150:341-351.
19. McMahon FJ, Buervenich S, Charney D, et al. Variation in
the gene
encoding the serotonin 2A receptor is associated with outcome
of antide-
pressant treatment. Am J Hum Genet. 2006;78:804-814.
20. Murphy GM, Kremer C, Rodrigues HE, Schatzberg AF.
Pharmacogenetics
of antidepressant medication intolerance. Am J Psychiatry.
2003;160:1830-
1835.
DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 75
P h a r m a c o l o g i c a l a s p e c t s
7 6
Pruebas farmacogenómicas en la práctica
clínica psiquiátrica
La incorporación en psiquiatría clínica de las prue-
bas farmacogenómicas ha ocurrido rápidamente en
los últimos siete años. Inicialmente se identificaron
genes de enzimas metabolizadoras de fármacos,
como el gen del citocromo P450 2D6. La tipificación
del gen del citocromo P450 2D6 que es altamente
variable da la oportunidad actualmente a los clíni-
cos de identificar a los metabolizadores pobres y los
ultrarrápidos para las sustancias que son sustrato
del 2D6. Con posterioridad se ha podido disponer
en la práctica clínica de genes que influyen en la
respuesta farmacodinámica de los medicamentos.
Entre los primeros “genes blanco” estuvo el gen del
transportador de serotonina (SLC6A4), el cual tiene
variantes que han demostrado que influyen en la
respuesta clínica de los pacientes con ancestros
europeos cuando son tratados con inhibidores
selectivos de la recaptura de serotonina. La tipifi-
cación de algunos de los genes del receptor de sero-
tonina también está disponible para guiar la prác-
tica clínica. La cuantificación de la utilidad clínica de
las pruebas farmacogenómicas está en desarrollo y
se han establecido las consideraciones éticas para
su realización. Considerando la cada vez más clara
costo-eficacia de la tipificación génica, reciente-
mente se ha pronosticado que las pruebas farma-
cogenómicas se solicitarán de rutina para orientar
la selección y dosificación de los fármacos psicotró-
picos.
Évaluation pharmacogénomique
psychiatrique en pratique clinique
L’évaluation pharmacogénomique psychiatrique
s’est rapidement imposée en pratique clinique au
cours de ces 7 dernières années. Les gènes d’en-
zymes métabolisant les médicaments, comme le
cytochrome P450 2D6 (CYP2D6), ont d’abord été
identifiés. Le génotypage de ce gène très variable
permet maintenant aux cliniciens d’identifier des
métaboliseurs lents et des métaboliseurs ultrara-
pides des substrats du 2D6. Des gènes influant sur
la réponse pharmacodynamique des médicaments
sont ensuite devenus disponibles en pratique cli-
nique. Parmi les premiers « gènes cibles », le gène
du transporteur de la sérotonine (SLC6A4) possède
des variants qui influent sur la réponse clinique des
patients d’ascendance européenne lorsqu’ils sont
traités avec des inhibiteurs sélectifs de la recapture
de la sérotonine. Le génotypage de certains gènes
du récepteur de la sérotonine est également dis-
ponible pour guider la réponse clinique. La quan-
tification de l’utilité clinique de l’évaluation phar-
macogénomique évolue et fait l’objet de
considérations éthiques. Il a été récemment prédit
qu’en raison de l’évidente rentabilité croissante du
génotypage, l’évaluation pharmacogénomique
devrait faire partie des examens de routine pour
sélectionner et ajuster la posologie des médica-
ments psychotropes.
21. Segman RH, Heresco-Levy U, Finkel B, et al. Association
between the
serotonin 2A receptor gene and tardive dyskinesia in chronic
schizophre-
nia. Mol Psychiatry. 2001;6:225-229.
22. Tan EC, Chong SA, Mahendran R, Dong F, Tan CH.
Susceptibility to neu-
roleptic-induced tardive dyskinesia and the T102C
polymorphism in the sero-
tonin type 2A receptor. Biol Psychiatry. 2001;50:144-147.
23. Lattuada E, Cavallaro R, Serretti A, Lorenzi C, Smeraldi E.
Tardive dyski-
nesia and DRD2, DRD3, DRD4, 5-HT2A variants in
schizophrenia: an associa-
tion study with repeated assessment. Int J Neuropsychopharmcol
2004;7:489-493.
24. Sodhi MS, Arranz MJ, Curtis D, et al. Association between
clozapine
response and allelic variation in the 5-HT2C receptor gene.
Neuroreport.
1995;7:169-172.
25. Arranz MJ, Munro J, Birkett J, et al. Pharmacogenetic
prediction of
clozapine response. Lancet. 2000;355:1615-1616.
26. Gunes A, Scordo MG, Jaanson P, Dahl ML. Serotonin and
dopamine
receptor gene polymorphisms and the risk of extrapyramidal
side effects in
perphenazine-treated schizophrenic patients.
Psychopharmacology
2007;190:479-484.
27. Reynolds GP, Zhang ZJ, Zhang XB. Association of
antipsychotic drug-
induced weight gain with a 5-HT2C receptor gene
polymorphism. Lancet.
2002;359:2086-2087.
28. Templeman LA, Reynolds GP, Arranz B, San L.
Polymorphisms of the 5-
HT2C receptor and leptin genes are associated with
antipsychotic drug-
induced weight gain in Caucasian subjects with a first-episode
psychosis.
Pharmacogenet Genomics. 2005;15:195-200.
29. De Luca V, Muller DJ, Hwang R, et al. HTR2C haplotypes
and antipsy-
chotics-induced weight gain: X-linked multimarker analysis.
Hum
Psychopharmacol. 2007;22:463-467.
30. Sallee FR, DeVane CL, Ferrell RE. Fluoxetine-related death
in a child with
cytochrome P-450 2D6 genetic deficiency. J Child Adolesc
Psychopharmacol.
2000;10:27-34.
31. Koski A, Ojanpera I, Vuori E, Sajantila A. A fatal doxepin
poisoning
associated with a defective CYP2D6 genotype. Am J Forensic
Med Pathol.
2007;28:259-261.
32. Stotland NL. Presidential address. Am J Psychiatry.
2009;166:1100-1104.
DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 76
E-Mail [email protected]
Editorial
Psychother Psychosom 2016;85:129–135
DOI: 10.1159/000443512
The Limitations of Genetic Testing in
Psychiatry
Steven L. Dubovsky
Department of Psychiatry, State University of New York at
Buffalo, Buffalo, N.Y. , and Departments of Psychiatry and
Medicine, University of Colorado, Denver, Colo. , USA
tocols for them [4] . In moderately differentiated breast
cancers, which comprise 50% of breast tumors, gene ex-
pression signatures for mitotic index, angiogenic poten-
tial, p53 mutational status, and estrogen and progester-
one dependence provide better stratification of prognosis
than histology [6] . However, despite such advances, there
is still not much clear integration between genomics and
clinical practice in oncology [7] .
Psychiatric Diagnosis
Numerous markers have been associated with psychi-
atric disorders, including genes for BDNF (brain-derived
neurotrophic factor), FOS (FBJ murine osteosarcoma vi-
ral oncogene homolog), COMT, DRD1, DRD2, DISC1,
GABABR1 (γ-aminobutyric acid B receptor 1), NR4A2
(nuclear receptor subfamily 4, group A, member 2),
ADORA2A (adenosine A2a receptor), CACNA1C (cal-
cium channel gene), sirtuin 1, LHPP, 5HTR1A, RNA-
binding proteins, and genes for myelination, glutaminer-
gic and GABAergic neurotransmission, oxidative stress,
signal transduction, response to the environment, cell
survival and proliferation, and cell shrinkage and apop-
tosis, among others [8–13] . Yet no genetic marker has yet
been shown to be useful in prospectively identifying any
specific psychiatric disorder [14] . Because genetic predis-
Despite the burgeoning number of psychiatric treat-
ments, we still do not know how to predict which one will
work best for which patient. The hope that genetic (single
gene effects) and genomic (multiple gene effects) testing
might be useful for diagnosis and treatment has been en-
couraged by decreased costs of genome sequencing and
studies demonstrating an association between mutations
in more than 3,000 genes and specific disease phenotypes
[1–3] . Are the data as promising in psychiatry as they are
in other fields?
Cancer Genomics
Genetic testing has been most promising in oncology.
For example, about 10% of cases of breast cancer have an
autosomal dominant pattern of transmission, most com-
monly mutations in the tumor suppressor genes BRCA1
and BRCA2 [4] . When BRCA1/2 mutations are found,
healthy women are offered a very close follow-up, as well
as prophylactic antiestrogen therapy or surgery, yet in
one study only 9.5% of high-risk women even underwent
genetic counseling, let alone testing [5] . Breast cancer risk
alleles have also been found for p53, PTEN (phosphate
and tensin homolog deleted from chromosome 10),
STK11, CDH1 and PALB2; however, these genetic factors
are rare, and there is not much research on screening pro-
Received: December 15, 2015
Accepted after revision: December 20, 2015
Published online: April 5, 2016
Steven L. Dubovsky, MD
Department of Psychiatry, University at Buffalo
462 Grider Street, Room 1182
Buffalo, NY 14215 (USA)
E-Mail dubovsky @ buffalo.edu
© 2016 S. Karger AG, Basel
0033–3190/16/0853–0129$39.50/0
www.karger.com/pps
http://dx.doi.org/10.1159%2F000443512
Dubovsky
Psychother Psychosom 2016;85:129–135
DOI: 10.1159/000443512
130
position in psychiatry is thinly distributed over thou-
sands of loci, each contributing a small effect, with con-
siderable overlap of brain systems and shared genetic fac-
tors [14] , sample size in most association studies has
generally been too small to produce meaningful, replica-
ble results [1, 15, 16] . In addition, epigenetic and oth-
er factors that alter DNA conformation can determine
whether susceptibility genes are expressed or suppressed
[10] , complicating analyses of the relationship between
genotype and phenotype. Even relevant genetic markers
can be difficult to interpret because inherited gene factors
appear to interact with each other and the environment
to contribute to both illness susceptibility and clinical
presentation [17, 18] .
Descriptive diagnoses in psychiatry have multiple do-
mains such as age of onset, constellations of specific
symptoms, functioning, comorbidity, and evolution over
time that assort differently in different patients in the
same category to produce functionally different condi-
tions [19] . Genetic profiles associated with any one of
these features are not likely to predict more global diag-
noses. If the direct path from genotype to phenotype ends
at discrete endophenotypes such as arousal, anhedonia,
information processing, stress responses, inflammation
and mood, rather than global diagnosis, attempts to link
the latter to specific genes are likely to prove frustrating
[20–23] , just as descriptive diagnoses in psychiatry do not
adequately consider important subtypes that exhibit dif-
ferent assortments of features such as age of onset, sever-
ity, progression or functioning.
Genetic Pharmacokinetic Studies
Since cytochrome P450 (CYP450) 1A2, 2D6, 2C9,
2C19, and 3A4 account for 60% of psychiatric drug me-
tabolism [24] , considerable interest has centered on using
the CYP450 genotype to predict response to psychotropic
medications [3] . However, genotype does not inevitably
predict phenotype because multiple copies of a more or
less active gene can result in more or less metabolic activ-
ity than would be expected from the allele that is identi-
fied. In addition, the metabolizer phenotype associated
with a particular genotype can be inhibited or enhanced
by a number of medications, substances, and foods [25–
30] . In an open study of 900 patients treated with venla-
faxine who were both genotyped and phenotyped for
CYP2D6, 4% were genotypically poor metabolizers, while
27% were phenotypically poor metabolizers, suggesting
that 23% of patients with other genotypes had converted
to a poor metabolizer phenotype as a result of concomi-
tant medications [31] .
Even if genotype inevitably predicted phenotype, the
correlation is stronger between CYP450 phenotype and
drug level than clinical response [32] , which is modified
by metabolism of most medications by more than one
enzyme, lack of linear kinetics and saturable elimination
for many drugs, unclear correlations between blood level
and response for many medications, and therapeutic win-
dows requiring therapeutic monitoring anyway for some
of them [24, 26, 33, 34] . Expression of CYP450 enzymes
in the brain, which influences drug effect, may be differ-
ent from their expression in the blood, or even in the in-
testine and liver [24] .
Drug Transporter Studies
Drug transporters, including P-glycoprotein (P-gp), or-
ganic ion transporters, and multidrug and toxin extrusion
proteins, modify the effect of CYP450 phenotype on drug
levels and drug action because they influence gastrointesti-
nal absorption, tissue uptake, and renal elimination as well
as transport in and out of the brain [35, 36] . In the iSPOT-
D (International Study to Predict Optimized Treatment in
Depression), two different MDR1 single nucleotide poly-
morphisms (SNPs) of the gene for P-gp (MDR1 or ABCB1)
were associated with better responses either to escitalo-
pram and sertraline or to venlafaxine, but there was no a
priori hypothesis, other relevant factors such as drug me-
tabolism, ethnicity, age, specific symptoms, or concomitant
illness were not addressed [36] , and DNA was collected af-
ter results were known rather than prospectively [37] .
Pharmacodynamic Studies
Both a deletion (short polymorphism or s-allele) and
an insertion (long polymorphism or l-allele) have been
found in the gene (SCL6A4) for the promoter region
(5-HTTLPR) of the gene for the serotonin transporter
(SERT) [32] . The short polymorphism (s) decreases and
the long polymorphism (l) increases SLC6A4 transcrip-
tion rates, resulting in less or more SERT expression, re-
spectively [32] . Research on the association of the s/s ge-
notype with a lower response rate to serotonin reuptake
inhibitors in some ethnic groups has been contradictory
[38–40] , and even without correction for multiple statisti-
cal tests, the SCL6A4 genotype explains at most 5% of the
variance in antidepressant response [30] . Attempts have
http://dx.doi.org/10.1159%2F000443512
Genetics in Psychiatry Psychother Psychosom 2016;85:129–
135
DOI: 10.1159/000443512
131
been made to correlate SNPs of the brain-specific voltage-
gated rectifier potassium channel (Kv11.1–3.1) and the
cardiac specific version (Kv11.1–1A) with risperidone
treatment response and changes in cardiac conduction,
respectively, in schizophrenia [41] , but the results have
not been robust.
Gene Network Studies
Because treatment outcome seems to be influenced by
multiple genetic polymorphisms, each with a small effect
[3, 21] , research has moved toward analysis of networks
of genes in the hope of developing more clinically useful
information [42–44] . However, such studies have not
produced clinically meaningful results [43, 44] . The Ge-
nome-Based Therapeutic Drugs for Depression (GEN-
DEP; n = 811) study, a substudy of the Sequenced Treat-
ment Alternatives to Relive Depression (STAR * D; n =
1,491) study, and the Munich Antidepressant Response
Study (n = 339), did not find any combination of genetic
markers that influenced treatment response in depres-
sion [1, 44] . Genome-wide association studies did not re-
veal any SNPs associated with response or remission of
nonbipolar, nonpsychotic, major depressive disorder
treated openly with serotonin reuptake inhibitors [45] ,
and the STAR * D study did not reveal any positive ge-
nome-wide association or top 25 SNP associations with
treatment response [45] . A genome-wide association
study from the Clinical Antipsychotic Trials of Interven-
tion Effectiveness (CATIE, n = 738) did not find any com-
binations of genetic markers that influenced treatment
response in schizophrenia [1, 44] .
Prospective Treatment Studies
Only a small number of reports have involved the pro-
spective use of genotyping to make treatment decisions.
An open study of 58 depressed inpatients reported that
genotyping for ABCB1 was associated with a shorter hos-
pital stay because patients with the TT/GG genotype were
more likely to have an increase in the dose of an antide-
pressant that was a P-gp substrate, although changing to
a non-P-gp substrate did not affect outcome [46] . The
study was not randomized, and numerous intervening
variables, including pharmacokinetics, comorbidity, and
history, were not considered.
Four studies have been supported by the manufactur-
er of a proprietary survey (GeneSight) of CYP2D6, 2C19,
2C9, and 1A2, SLC6A4, and 5HTR2A genotypes that gen-
erates a ‘composite report’ classifying antidepressants
and antipsychotic drugs used in the treatment of depres-
sion into three categories: ‘use as directed’, ‘use with cau-
tion’, and ‘use with caution and with more frequent mon-
itoring’. An 8-week open study of 44 patients assigned in
a nonrandom manner to treatment guided by the com-
posite report (guided treatment) or nonguided treatment
by the same clinicians, who were involved with the prod-
uct, reported that patients in the guided group were less
likely to receive medications in the ‘use with caution and
with more frequent monitoring’ category, presumably
because of reluctance by the guided clinicians to prescribe
medications that required more monitoring [47] . Al-
though improvement of depression was similar for the
first 4 weeks in both groups, a single measure at 8 weeks
indicated increased depression scores for the nonguided
but not the guided group. No explanation was offered for
the final increase in depression scores in the nonguided
group, when multiple earlier ratings demonstrated a
steady decrease in scores. Improvement in the guided
group was not impressive, and it is impossible to know
whether comorbid factors, concomitant medications,
treatment adherence, patient enthusiasm, substance use,
adjunctive psychotherapy, clinician knowledge of treat-
ment condition, and open ratings affected the conduct of
treatment or the outcome assessment.
A second open, nonrandomized study conducted by
the same group in 227 mildly-moderately depressed pa-
tients, 165 of whom completed 8 weeks of treatment, re-
ported that patients in the guided group were twice as
likely to respond [48] . Since clinicians reported substan-
tial levels of confidence in the genetic reports, it is possi-
ble that they worked more vigorously with patients in the
guided group, that patients in the guided group were
more adherent with a treatment approach they thought
would be more effective, or that they reported better re-
sults to please the investigators.
In a double-blind, randomized, controlled trial of
GeneSight, 25 depressed patients were assigned to treat-
ment as usual and 26 to guided treatment [49] . Improve-
ment was numerically greater in patients in the guided
than in the treatment-as-usual group, but none of the
group differences were statistically significant. In a fourth
report from the same company [50] , 97 patients with a
depressive or anxiety disorder treated openly by a single
psychiatrist with one of the medications in the genetic sur-
vey were followed openly for 1 year. The 9 patients taking
at least one medication in the ‘use with caution’ category
had significantly more total health care visits and nonpsy-
http://dx.doi.org/10.1159%2F000443512
Dubovsky
Psychother Psychosom 2016;85:129–135
DOI: 10.1159/000443512
132
chiatric medical visits than the other subjects and had a
higher average cost of care. However, these patients also
took more medications than the other subjects, and there
was a significant correlation between the number of med-
ications taken and the two outcome variables. When dif-
ferent statistical analyses were performed on the same
data (e.g. analysis of variance and t tests), some were sig-
nificant and some were not, and no correction was made
for multiple statistical tests. Although the authors con-
tended that their genetic analysis could save health care
costs, this hypothesis was not actually tested. Since no data
were available on medical comorbidity and severity of
psychiatric illness, the possibility was not considered that
the small number of patients in the ‘use with caution’ cat-
egory had more health care visits and took more medi-
cations because they were sicker psychiatrically or medi-
cally.
Gene Expression
Current genotyping approaches in psychiatry consider
the presence or absence of a particular allele or group of
alleles, but expression of those genes may be suppressed,
modified, or enhanced by a number of factors, including
circadian transcription patterns, epistasis (gene interac-
tions), regulatory regions, epigenetic factors, and noncod-
ing RNA [14] . Histone modification and DNA methyla-
tion in response to experience, inflammation, the illness,
and the medications used to treat it can induce or suppress
multiple genes, and genotype itself can affect methylation
of regulatory sites that leads to epigenetic changes in brain
development [51] . Micro-RNAs and short interfering
RNAs are short, noncoding posttranslational regulators of
gene expression that target hundreds of mRNA transcripts
to influence gene networks [27, 52, 53] . The expression of
genes for CYP450 enzymes is altered by promoter meth-
ylation, micro-RNAs associated with inflammation and
other illnesses [27] , and some medications [54] , resulting
in an altered CYP450 phenotype.
Limitations of Pharmacogenetic Testing in
Psychiatry
No matter how much we may want to translate direct-
ly to clinical diagnosis reports of an association between
a diagnosis and a genetic marker, the nature of the current
level of knowledge does not permit this application. Sam-
ple sizes in most existing studies have been too small to
produce meaningful, replicable results because of the
clinical and genetic heterogeneity of psychiatric disorders
[55] , and the combined influence of multiple genes, each
with a small effect size [1, 15, 16] . Most studies have uti-
lized retrospective or post hoc analyses rather than pro-
spective a priori hypotheses [56] , and statistical signifi-
cance is often inflated by lack of correction for multiple
statistical tests [16] . The majority of studies lack replica-
tion in independent samples, especially by different in-
vestigators [16] . Even robust findings would not be clini-
cally applicable until a prospective study demonstrated
their ability to preferentially predict one diagnosis or even
clinically relevant feature over another.
Using genotype to predict response to medications is
even more problematic. Pharmacogenetic studies have
been conducted in normal subjects or patients who are
not taking other medications and who do not have other
illnesses, limiting extrapolation to most clinical settings
[33, 34] . Most studies do not control for the effect on the
expression of CYP450 and other genes of age [27] , ethnic-
ity [30] , smoking [30, 34] , and use of substances such as
alcohol, hormones, St. John’s wort, caffeine, cabbage, and
grapefruit juice [30] . Genetic studies of treatment out-
come have not measured nonadherence [57] , but as the
rate of nonadherence increases in any population, statis-
tical power to detect a genotype effect decreases substan-
tially [1] . For medications that are chiral mixtures of en-
antiomers with different actions, the metabolism of each
enantiomer may be by different enzymes [58] . Active me-
tabolites with their own metabolic pathways may en-
hance or interfere with therapeutic or toxic effects pre-
dicted by the presumed metabolism of the parent drug
[59, 60] . In most instances, more than one genetic factor
affects drug levels and disposition [3] , and interactions
between these factors can be difficult to predict.
A clear demonstration of a genotype/blood level rela-
tionship in a single dose or 8-week study may not corre-
late with chronic treatment, in which compensatory
changes in secondary metabolic pathways and drug trans-
porters, gene up- or downregulation, saturation pharma-
cokinetics and other factors may modify the impact of
oxidative enzyme polymorphisms on final drug level [24,
59] . With chronic treatment, some psychotropic drug
metabolites form complexes with P450 enzymes that alter
or even reverse the acute effect on metabolism [25] . Long-
term changes in P450 enzymes also occur in the brain,
with further unpredictable effects, not only on the sub-
strate drug, but on neurotransmitters and neurosteroids
metabolized by the same enzymes on which the medica-
tion may act [25] . Another complicating factor is that the
http://dx.doi.org/10.1159%2F000443512
Genetics in Psychiatry Psychother Psychosom 2016;85:129–
135
DOI: 10.1159/000443512
133
illness, as well as medications used to treat it, can alter the
relationship between pharmacologic genotype and phe-
notype. For example, many proinflammatory cytokines
and acute-phase proteins that are associated with mood
and anxiety disorders [61] act on transcription or post-
translational protein modification to downregulate some
CYP450 genes and upregulate others [62] . At the same
time, suppression of cytokines by antidepressants can al-
ter gene expression in directions that antagonize im-
provement of depression [25] . The impact of evolution of
the illness and its response to different treatments in
modifying therapeutic strategies during the course of
treatment of cancer is relatively straightforward to study
by virtue of methodologies for examining genotype and
phenotype of cellular clones, but it is still difficult to de-
velop the correct approach to well-characterized tumors
[63] . The absence of such measures in psychiatric diagno-
ses makes this prospect considerably more difficult.
Where Do We Go from Here?
Psychiatrists, whose work frequently involves ambigu-
ous clinical problems, and who must often consider con-
tradictory elements of patient presentations and avoid
premature closure, can have a remarkably low tolerance
for ambiguity, conflict, and delayed gratification when it
comes to the latest laboratory study. The hope that phar-
macogenetic testing will result in unambiguous ‘person-
alized psychiatry’ should not lead to quick adoption of
technologies that have not yet been demonstrated to reli-
ably predict a specific course or a need for a specific med-
ication, the choice of which remains largely empirical. Af-
ter all, genetic associations are statistical, but medical
practice is personal [14] . Yet there is tremendous pressure
to translate each new report of such associations to our
patients, not only from our own need to appear ‘scien-
tific’ and from industry marketing of proprietary tests,
but from the marketing of ideas by thought leaders with
an intellectual attachment to the latest conceptualization
of genetic causality [64] .
It is a continuing challenge to examine new genetic
findings critically without applying them immediately in
the clinic. When adequately powered studies that address
gene number and expression and that control for real-life
factors that affect outcome such as comorbidity, poly-
pharmacy, environmental exposure, age, gender, ethnic-
ity, substance use, and treatment adherence emerge [44] ,
clinicians who have not put new information into action
before integrating it with emerging knowledge about di-
agnosis, neurobiology, and the evolution of complex dis-
orders will be ready to apply them effectively.
Disclosure Statement
Dr. Dubovsky has received research support from Janssen, Ot-
suka, Sumitomo, Neurocrine, Tower Foundation, Wendt
Founda-
tion, Oshei Foundation and Patrick Lee Foundation. The author
has no other conflicts of interest to disclose.
References
1 Malhotra AK, Zhang J-P, Lencz T: Pharmaco-
genetics in psychiatry: translating research
into clinical practice. Mol Psychiatry 2012; 17:
760–769.
2 Cox DBT, Platt RJ, Zhang F: Therapeutic ge-
nome editing: prospects and challenges. Nat
Med 2015; 21: 121–131.
3 Trembplay J, Hamet P: Role of genomics on
the path to personalized medicine. Metabo-
lism 2013; 62:S2–S5.
4 Stuckey AR, Onstad MA: Hereditary breast
cancer: an update on risk assessment and ge-
netic testing in 2015. Am J Obstet Gynecol
2015; 213: 161–165.
5 Bellcross CA, Peipins LA, McCarty FA, Ro-
driguez JL, Hawkins NA, Hensley Alford S,
Leadbetter S: Characteristics associated with
genetic counseling referral and BRCA1/2 test-
ing among women in a large integrated health
system. Genet Med 2015; 17: 43–50.
6 Ivshina AV, George J, Senko O, Mow B, Putti
TC, Smeds J, Lindahl T, Pawitan Y, Hall P,
Nordgren H, Wong JEL, Liu ET, Bergh J,
Kuznetsove VA, Miller LD: Genetic reclassi-
fication of histologic grade delineates new
clinical subtypes of breast cancer. Cancer Res
2006; 66: 10292–10301.
7 Anonymous: The future of cancer genomics.
Nat Med 2015; 21: 99.
8 Le-Niculescu H, Balaraman Y, Patel SD,
Ayalew M, Gupta J, Kuczenski R, Shekhar A,
Shekhar A, Schork N, Geyer MA, Niculescu
AB: Convergent functional genomics of anxi-
ety disorders: translational identification of
genes, biomarkers, pathways and mecha-
nisms. Transl Psychiatry 2011; 1:e9.
9 Carrard A, Salzmann A, Malafosse A, Karege
F: Increased DNA methylation status of the
serotonin receptor 5HTR1A gene promoter
in schizophrenia and bipolar disorder. J Af-
fect Disord 2011; 132: 450–453.
10 Mill J, Tang T, Kaminsky Z, Khare T, Yaz-
danpanah S, Bouchard L, Jia P, Assadzadeh A,
Flanagan J, Schumacher A, Wang SC, Petro-
nis A: Epigenomic profiling reveals DNA-
methylation changes associated with major
psychosis. Am J Hum Genet 2008; 82: 696–
711.
11 Fullerton JM, Tiwari Y, Agahi G, Heath A,
Berk M, Mitchell PB, Schofield PR: Assessing
oxidative pathway genes as risk factors for bi-
polar disorder. Bipolar Disord 2010; 12: 550–
556.
12 Hoenicka J, Garrido E, Ponce G, Rodriguez-
Jimenez R, Martinez I, Rubio G, Jimenez-Ar-
riero MA, Palomo T: Sexually dimorphic in-
teraction between the DRD1 and COMT
genes in schizophrenia. Am J Med Genet
2010; 153B:984–954.
http://dx.doi.org/10.1159%2F000443512
Dubovsky
Psychother Psychosom 2016;85:129–135
DOI: 10.1159/000443512
134
13 Hong CJ, Liou YJ, Bai Y, Chen TT, Wang Y,
Tsai SJ: Dopamine receptor D2 gene is associ-
ated with weight gain in schizophrenia pa-
tients under long-term atypical antipsychotic
treatment. Pharmacogenet Genomics 2010;
20: 359–366.
14 Li JZ: Circadian rhythms and mood: opportu-
nities for multi-level analyses in genomics
and neuroscience. Bioessays 2013; 36: 305–
315.
15 Genetics of Personality Consortium: Meta-
analysis of genome-wide association studies
for neuroticism, and the polygenic associa-
tion with major depression. JAMA Psychiatry
2015; 72: 642–650.
16 Gillis NK, Innocenti F: Evidence required to
demonstrate clinical utility of pharmacoge-
netic testing: the debate continues. Clin Phar-
macol Ther 2014; 96: 655–657.
17 Fanous AH, Kendler KS: Genetic heterogene-
ity, modifier genes, and quantitative pheno-
types in psychiatric illness: searching for a
framework. Mol Psychiatry 2005; 10: 6–13.
18 Patel SD, Le-Niculescu H, Koller DL, Green
SD, Lahiri DK, McMahon FJ, Nurnberger JIJ,
Niculescu AB: Coming to grips with complex
disorders: genetic risk prediction in bipolar
disorder using panels of genes identified
through convergent functional genomics. Am
J Med Genet 2010; 153B:850–877.
19 Fava GA, Guidi J, Grandi S, Hasler G: The
missing link between clinical states and bio-
markers in mental disorders. Psychother Psy-
chosom 2014; 83: 136–141.
20 Van Borkulo C, Boschioo L, Borsboom D,
Pennix BWJH, Waldorp LJ, Schoevers RA:
Association of symptom network structure
with the course of longitudinal depression.
JAMA Psychiatry 2015; 72: 1219–1226.
21 Keers R, Aitchison KJ: Pharmacokinetics of
antidepressant response. Expert Rev Neu-
rother 2011; 11: 101–125.
22 Domschke K: Clinical and molecular genetics
of psychotic depression. Schizophr Bull 2013;
39: 766–775.
23 Fabbri C, Porcelli S, Serretti A: From pharma-
cogenetics to pharmacogenomics: the way to-
ward the personalization of antidepressant
treatment. Can J Psychiatry 2014; 59: 62–75.
24 Preissner SC, Hoffmann MF, Preissner R,
Dunkel M, Gewiess A, Preissner S: Polymor-
phic cytochrome P450 enzymes (CYPs) and
their role in personalized therapy. PLoS One
2013; 8:e82562.
25 Wladyslawa AD: The influence of long-term
treatment with psychotropic drugs on cyto-
chrome P450: the involvement of different
mechanisms. Expert Opin Drug Metab Toxi-
col 2005; 1: 203–217.
26 Crews KR, Gaedigk A, Dunnenberger HM,
Leeder JS, Klein TE, Caudle KE, Haidar CE,
Shen DD, Callaghan JT, Sadhasivam S, Prows
CA, Kharasch ED, Skaar TC: Clinical Phar-
macogenetics Implementation Consortium
guidelines for cytochrome P450 2D6 geno-
type and codeine therapy: 2014 update. Clin
Pharmacol Ther 2014; 95: 376–382.
27 Zanger UM, Klein K, Thomas M, Rieger JK,
Tremmel R, Kandel BA, Klein BA, Klein M,
Magdy T: Genetics, epigenetics, and regulation
of drug-metabolizing cytochrome p450 en-
zymes. Clin Pharmacol Ther 2014; 95: 258–261.
28 Misaka S, Kawabe K, Onoue S, Werba JP, Gi-
roli M, Tamaki S, Kan T, Kimura J, Watanabe
H, Yamada S: Effects of green tea catechins on
cytochrome P450 2B6, 2C8, 2C19, 2D6 and
3A activities in human liver and intestinal mi-
crosomes. Drug Metab Pharmacokinet 2013;
28: 244–249.
29 Yamasaki I, Yamada M, Uotsu N, Teramoto
S, Takayanagi R, Yamada Y: Inhibitory effects
of kale ingestion on metabolism by cyto-
chrome P450 enzymes. Biomed Res 2012; 33:
235–242.
30 Jones DS, Perlis RH: Pharmacogenetics, race,
and psychiatry: prospects and challenges.
Harvard Rev Psychiatry 2006; 14: 92–108.
31 Preskorn SH, Kane CP, Lobello K, Nichols AI,
Fayyad R, Buckley G, Focht K, Guico-Pabia
CJ: Cytochrome P450 2D6 phenoconversion
is common in patients being treated for de-
pression: implications for personalized medi-
cine. J Clin Psychiatry 2013; 74: 614–621.
32 Alter CN, Hornberger J, Shewade A, Cruz V,
Garrison J, Mrazek D: Clinical validity of cy-
tochrome P450 metabolism and serotonin
gene variants in psychiatric pharmacothera-
py. Int Rev Psychiatry 2013; 25: 509–533.
33 De Leon J, Arranz GR: Pharmacogenetic test-
ing in psychiatry: a review of features and clin-
ical realities. Clin Lab Med 2008; 28: 599–617.
34 Kirchheiner J, Brosen K, Dahl ML, Gram LF,
Kasper S, Roots I, Sjoqvist F, Spina E, Brock-
moller J: CYP2D6 and CYP2C19 genotype-
based dose recommendations for antidepres-
sants: a first step towards subpopulation-spe-
cific dosages. Acta Psychiatr Scand 2001; 104:
173–192.
35 Rosenhagen MC, Uhr M: The clinical impact
of ABCB1 polymorphisms in the treatment of
psychiatric diseases. Curr Pharm Des 2011;
17: 2843–2851.
36 Goswami S, Yee SW, Stocker S, Mosley JD,
Kubo M, Castro R, Mefford JA, Wen C, Liang
X, Witte J, Brett C, Maeda S, Simpson MD,
Hedderson MM, Davis RL, Roden DM, Gia-
comini KM, Savic RM: Genetic variants in
transcription factors are associated with the
pharmacokinetics and pharmacodynamics of
metformin. Clin Pharmacol Ther 2014; 96:
370–370.
37 Schatzberg AF, DeBattista C, Lazzeroni LC,
Etkin A, Murphy GM, Williams LM: ABCB1
genetic effects on antidepressant outcomes: a
report from the iSPOT-D trial. Am J Psychia-
try 2015; 172: 751–759.
38 Porcelli S, Fabbri C, Serretti A: Meta-analysis
of serotonin transporter gene promoter poly-
morphism (5-HTTLPR) association with an-
tidepressant efficacy. Eur Neuropsychophar-
macol 2012; 22: 239–258.
39 Taylor MJ, Sen S, Bhagwagar Z: Antidepres-
sant response and the serotonin transporter-
linked polymorphic region. Biol Psychiatry
2010; 68: 536–543.
40 Kang HJ, Kim JM, Stewart R, Kim SY, Bae KY,
Kim SW, Shin MG, Yoon JS: Association of
SLC6A4 methylation with early adversity,
characteristics and outcomes in depression.
Prog Neuropsychopharmacol Biol Psychiatry
2013; 44: 23–28.
41 Heide J, Zhang F, Bigos KL, Mann SA, Carr
VJ, Weickert CS, Green MJ, Weinberger
DR, Vandenberg JI: Differential response to
risperidone in schizophrenia patients by
KCNH2 genotype and drug metabolizer sta-
tus. Am J Psychiatry 2016; 173: 53–59.
42 Costa e Silva JA: Personalized medicine in
psychiatry: new technologies and approaches.
Metabolism 2013; 62:S40–S44.
43 Singh AB, Bousman CA, Ng C, Berk M: Anti-
depressant pharmacogenetics. Curr Opin
Psychiatry 2014; 27: 43–51.
44 Hamilton SP: The promise of psychiatric
pharmacogenomics. Biol Psychiatry 2015; 77:
29–35.
45 Ji Y, Biernacka JM, Hebbring S, Jenkins GD,
Batzler A, Snyder KA, Drews MS, Desta Z,
Flockart D, Mushiroda T, Kubo M, Naka-
mura Y, Kamatani N, Schaid D, Weinshil-
boum RM, Mrazek D: Pharmacogenomics
of selective serotonin reuptake inhibitor
treatment for major depressive disorder: ge-
nome-wide associations and functional ge-
nomics. Pharmacogenomics J 2013; 13: 456–
463.
46 Breitenstein B, Scheuer S, Pfister H, Uhr M,
Lucae S, Holsboer F, Ising M, Bruckl T: The
clinical application of ABCB1 genotyping in
antidepressant treatment: a pilot study. CNS
Spectr 2014; 19: 165–175.
47 Hall-Flavin DK, Winner JG, Allen J, Jordan JJ,
Nesheim RS, Snyder KA, Drews MS, Eister-
hold LL, Biernacka JM, Mrazek D: Using a
pharmacogenomic algorithm to guide the
treatment of depression. Transl Psychiatry
2012; 2:e172.
48 Hall-Flavin DK, Winner JG, Allen J, Carhart
JM, Proctor B, Snyder KA, Drews MS, Eister-
hold LL, Geske J, Mrazek D: Utility of inte-
grated pharmacogenomic testing to support
the treatment of major depressive disorder in
a psychiatric outpatient setting. Pharmaco-
genet Genomics 2013; 23: 535–548.
49 Winner JG, Carhart JM, Alter CN, Allen J,
Dechairo BM: A prospective, randomized,
double-blind study assessing the clinical im-
pact of integrated pharmacogenomic testing
for major depressive disorder. Discov Med
2013; 16: 219–227.
50 Winner JG, Allen J, Altar CA, Spahic-Miha-
jlovic A: Psychiatric pharmacogenomics pre-
dicts health resource utilization of outpatients
with anxiety and depression. Transl Psychia-
try 2013; 3:e242.
http://dx.doi.org/10.1159%2F000443512
Genetics in Psychiatry Psychother Psychosom 2016;85:129–
135
DOI: 10.1159/000443512
135
51 Hannon E, Spiers H, Viana J, Pidsley R, Bur-
rage J, Murphy TM, Troakes C, Turecki G,
O’Donovan MC, Schalkwyk LC, Bray NJ, Mill
J: Methylation QTLS in the developing brain
and their enrichment in schizophrenia risk
loci. Nat Neurosci 2016; 19: 48–54.
52 Geaghan M, Cairns MJ: MicroRNA and post-
transcriptional dysregulation in psychiatry.
Biol Psychiatry 2015; 78: 231–239.
53 Xu Q, Wu X, Xiong Y, Xing Q, He L, Qin S:
Pharmacogenomics can improve antipsy-
chotic treatment in schizophrenia. Front Med
2013; 7: 180–190.
54 Gardiner E, Carroll A, Tooney PA, Cairns MJ:
Antipsychotic drug-associated gene-miRNA
interaction in T-lymphocytes. Int J Neuro-
psychopharmacol 2014; 17: 929–943.
55 Le-Niculescu H, Kurian SM, Yehyawi N, Dike
C, Patel SD, Edenberg HJ, Tsuang MT, Salo-
mon DR, Nurnberger JIJ, Niculescu AB: Iden-
tifying blood biomarkers for mood disorders
using convergent functional genomics. Mol
Psychiatry 2009; 14: 156–174.
56 Janssens ACJW, Deverka PA: Useless until
proven effective: the clinical utility of pre-
emptive pharmacogenetic testing. Clin Phar-
macol Ther 2014; 96: 652–654.
57 Melfi CA, Chawla AJ, Croghan TW, Hanna
MP, Kennedy S, Sredl K: The effects of adher-
ence to antidepressant treatment guidelines
on relapse and recurrence of depression. Arch
Gen Psychiatry 1998; 55: 1128–1132.
58 Crettol S, de Leon J, Hiemke C, Eap CB: Phar-
macogenomics in psychiatry: from therapeu-
tic drug monitoring to genomic medicine.
Clin Pharmacol Ther 2014; 95: 254–257.
59 Kirchheiner J, Nickchen K, Bauer M, Wong
M-L, Roots I, Brockmoller J: Pharmacogenet-
ics of antidepressants and antipsychotics: the
contribution of allelic variations to the phe-
notype of drug response. Mol Psychiatry
2004; 9: 442–473.
60 Seeringer A, Kirchheiner J: Pharmacogenet-
ics-guided dose modifications of antidepres-
sants. Clin Lab Med 2008; 28: 619–626.
61 Young JJ, Bruno D, Pomara N: A review of the
relationship between proinflammatory cyto-
kines and major depressive disorder. J Affect
Disord 2014; 169: 15–20.
62 Harvey RD, Morgan ET: Cancer, inflamma-
tion, and therapy: effects on cytochrome
p450-mediated drug metabolism and impli-
cations for novel immunotherapeutic agents.
Clin Pharmacol Ther 2014; 96: 449–457.
63 Michor F, Beal K: Improving cancer treat-
ment via mathematical modeling: surmount-
ing the challenges is worth the effort. Cell
2015; 163: 1059–1063.
64 Dubovsky SL, Dubovsky AN: Psychotropic
Drug Prescriber’s Survival Guide: Ethical Pre-
scribing in the Age of Big Pharma. New York,
Norton, 2007.
http://dx.doi.org/10.1159%2F000443512
CitRef_1: CitRef_2: CitRef_3: CitRef_4: CitRef_5: CitRef_6:
CitRef_7: CitRef_8: CitRef_9: CitRef_10: CitRef_11:
CitRef_12: CitRef_13: CitRef_14: CitRef_15: CitRef_16:
CitRef_17: CitRef_18: CitRef_19: CitRef_20: CitRef_21:
CitRef_22: CitRef_23: CitRef_24: CitRef_25: CitRef_26:
CitRef_27: CitRef_28: CitRef_29: CitRef_30: CitRef_31:
CitRef_32: CitRef_33: CitRef_34: CitRef_35: CitRef_36:
CitRef_37: CitRef_38: CitRef_39: CitRef_40: CitRef_41:
CitRef_42: CitRef_43: CitRef_44: CitRef_45: CitRef_46:
CitRef_47: CitRef_48: CitRef_49: CitRef_50: CitRef_51:
CitRef_52: CitRef_53: CitRef_54: CitRef_55: CitRef_56:
CitRef_57: CitRef_58: CitRef_59: CitRef_60: CitRef_61:
CitRef_62: CitRef_63:

Contenu connexe

Similaire à linical pharmacogenomics consists of the appli-cation of res.docx

DDS personalised medicines M.Pharma 1st Sem Pharmaceutics.pptx
DDS personalised medicines M.Pharma 1st Sem Pharmaceutics.pptxDDS personalised medicines M.Pharma 1st Sem Pharmaceutics.pptx
DDS personalised medicines M.Pharma 1st Sem Pharmaceutics.pptxkushaltegginamani18
 
Pharmacogenetics- Introduction, History with case Study
Pharmacogenetics- Introduction, History with case StudyPharmacogenetics- Introduction, History with case Study
Pharmacogenetics- Introduction, History with case StudyPushpak Bhati
 
Pharmacogenomics: The right drug to the right person.
Pharmacogenomics: The right drug to the right person.Pharmacogenomics: The right drug to the right person.
Pharmacogenomics: The right drug to the right person.University of Allahabad
 
clinical._pharmacology._ESSAY
clinical._pharmacology._ESSAYclinical._pharmacology._ESSAY
clinical._pharmacology._ESSAYDimitrios Brachos
 
PERSONALIZED MEDICINE AND PHARMACOGENETICS
PERSONALIZED MEDICINE  AND PHARMACOGENETICSPERSONALIZED MEDICINE  AND PHARMACOGENETICS
PERSONALIZED MEDICINE AND PHARMACOGENETICSAravindgowda6
 
The-Case-for-Practice-Integration[1]
The-Case-for-Practice-Integration[1]The-Case-for-Practice-Integration[1]
The-Case-for-Practice-Integration[1]Amy Williams
 
Soal dan Pembahasan Farmakogenomik dan Personalized Medicine
Soal dan Pembahasan Farmakogenomik dan Personalized MedicineSoal dan Pembahasan Farmakogenomik dan Personalized Medicine
Soal dan Pembahasan Farmakogenomik dan Personalized MedicineNesha Mutiara
 
Pharmacogenomics.pptx
Pharmacogenomics.pptxPharmacogenomics.pptx
Pharmacogenomics.pptxAnusha K R
 
WHMS PGx Presentation
WHMS PGx PresentationWHMS PGx Presentation
WHMS PGx PresentationOrion Cuffe
 
Target discovery and validation
Target discovery and validation Target discovery and validation
Target discovery and validation ANAND SAGAR TIWARI
 
AkrodouY-Week7 Project Part 2a
AkrodouY-Week7 Project Part 2aAkrodouY-Week7 Project Part 2a
AkrodouY-Week7 Project Part 2aYawo Akrodou
 
Genetics of antipsychotic drug outcome and implications for the clinician in...
Genetics of antipsychotic drug outcome and implications for the clinician  in...Genetics of antipsychotic drug outcome and implications for the clinician  in...
Genetics of antipsychotic drug outcome and implications for the clinician in...BARRY STANLEY 2 fasd
 
Session 1 part 1
Session 1 part 1Session 1 part 1
Session 1 part 1plmiami
 
Pharmacoepidemiology
PharmacoepidemiologyPharmacoepidemiology
PharmacoepidemiologyStanley Palma
 
Main PostPharmacokinetics and PharmacodynamicsAs advance.docx
Main PostPharmacokinetics and PharmacodynamicsAs advance.docxMain PostPharmacokinetics and PharmacodynamicsAs advance.docx
Main PostPharmacokinetics and PharmacodynamicsAs advance.docxwkyra78
 
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics Pharmacogenomics, Pharmacogenetics and Pharmacokinetics
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics Zohaib HUSSAIN
 

Similaire à linical pharmacogenomics consists of the appli-cation of res.docx (20)

DDS personalised medicines M.Pharma 1st Sem Pharmaceutics.pptx
DDS personalised medicines M.Pharma 1st Sem Pharmaceutics.pptxDDS personalised medicines M.Pharma 1st Sem Pharmaceutics.pptx
DDS personalised medicines M.Pharma 1st Sem Pharmaceutics.pptx
 
Pharmacogenetics- Introduction, History with case Study
Pharmacogenetics- Introduction, History with case StudyPharmacogenetics- Introduction, History with case Study
Pharmacogenetics- Introduction, History with case Study
 
Pharmacogenomics: The right drug to the right person.
Pharmacogenomics: The right drug to the right person.Pharmacogenomics: The right drug to the right person.
Pharmacogenomics: The right drug to the right person.
 
clinical._pharmacology._ESSAY
clinical._pharmacology._ESSAYclinical._pharmacology._ESSAY
clinical._pharmacology._ESSAY
 
PERSONALIZED MEDICINE AND PHARMACOGENETICS
PERSONALIZED MEDICINE  AND PHARMACOGENETICSPERSONALIZED MEDICINE  AND PHARMACOGENETICS
PERSONALIZED MEDICINE AND PHARMACOGENETICS
 
pharmacogenomics
pharmacogenomicspharmacogenomics
pharmacogenomics
 
The-Case-for-Practice-Integration[1]
The-Case-for-Practice-Integration[1]The-Case-for-Practice-Integration[1]
The-Case-for-Practice-Integration[1]
 
Soal dan Pembahasan Farmakogenomik dan Personalized Medicine
Soal dan Pembahasan Farmakogenomik dan Personalized MedicineSoal dan Pembahasan Farmakogenomik dan Personalized Medicine
Soal dan Pembahasan Farmakogenomik dan Personalized Medicine
 
Personalized medicine
Personalized medicinePersonalized medicine
Personalized medicine
 
OBJECTIVE 3.pptx
OBJECTIVE 3.pptxOBJECTIVE 3.pptx
OBJECTIVE 3.pptx
 
Pharmacogenomics.pptx
Pharmacogenomics.pptxPharmacogenomics.pptx
Pharmacogenomics.pptx
 
Genomics and proteomics
Genomics and proteomicsGenomics and proteomics
Genomics and proteomics
 
WHMS PGx Presentation
WHMS PGx PresentationWHMS PGx Presentation
WHMS PGx Presentation
 
Target discovery and validation
Target discovery and validation Target discovery and validation
Target discovery and validation
 
AkrodouY-Week7 Project Part 2a
AkrodouY-Week7 Project Part 2aAkrodouY-Week7 Project Part 2a
AkrodouY-Week7 Project Part 2a
 
Genetics of antipsychotic drug outcome and implications for the clinician in...
Genetics of antipsychotic drug outcome and implications for the clinician  in...Genetics of antipsychotic drug outcome and implications for the clinician  in...
Genetics of antipsychotic drug outcome and implications for the clinician in...
 
Session 1 part 1
Session 1 part 1Session 1 part 1
Session 1 part 1
 
Pharmacoepidemiology
PharmacoepidemiologyPharmacoepidemiology
Pharmacoepidemiology
 
Main PostPharmacokinetics and PharmacodynamicsAs advance.docx
Main PostPharmacokinetics and PharmacodynamicsAs advance.docxMain PostPharmacokinetics and PharmacodynamicsAs advance.docx
Main PostPharmacokinetics and PharmacodynamicsAs advance.docx
 
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics Pharmacogenomics, Pharmacogenetics and Pharmacokinetics
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics
 

Plus de croysierkathey

1.  Discuss the organization and the family role in every one of the.docx
1.  Discuss the organization and the family role in every one of the.docx1.  Discuss the organization and the family role in every one of the.docx
1.  Discuss the organization and the family role in every one of the.docxcroysierkathey
 
1.  Compare and contrast DEmilios Capitalism and Gay Identity .docx
1.  Compare and contrast DEmilios Capitalism and Gay Identity .docx1.  Compare and contrast DEmilios Capitalism and Gay Identity .docx
1.  Compare and contrast DEmilios Capitalism and Gay Identity .docxcroysierkathey
 
1.Purpose the purpose of this essay is to spread awareness .docx
1.Purpose the purpose of this essay is to spread awareness .docx1.Purpose the purpose of this essay is to spread awareness .docx
1.Purpose the purpose of this essay is to spread awareness .docxcroysierkathey
 
1.  Tell us why it is your favorite film.2.  Talk about the .docx
1.  Tell us why it is your favorite film.2.  Talk about the .docx1.  Tell us why it is your favorite film.2.  Talk about the .docx
1.  Tell us why it is your favorite film.2.  Talk about the .docxcroysierkathey
 
1.What are the main issues facing Fargo and Town Manager Susan.docx
1.What are the main issues facing Fargo and Town Manager Susan.docx1.What are the main issues facing Fargo and Town Manager Susan.docx
1.What are the main issues facing Fargo and Town Manager Susan.docxcroysierkathey
 
1.Writing Practice in Reading a PhotographAttached Files.docx
1.Writing Practice in Reading a PhotographAttached Files.docx1.Writing Practice in Reading a PhotographAttached Files.docx
1.Writing Practice in Reading a PhotographAttached Files.docxcroysierkathey
 
1.Some say that analytics in general dehumanize managerial activitie.docx
1.Some say that analytics in general dehumanize managerial activitie.docx1.Some say that analytics in general dehumanize managerial activitie.docx
1.Some say that analytics in general dehumanize managerial activitie.docxcroysierkathey
 
1.What is the psychological term for the symptoms James experiences .docx
1.What is the psychological term for the symptoms James experiences .docx1.What is the psychological term for the symptoms James experiences .docx
1.What is the psychological term for the symptoms James experiences .docxcroysierkathey
 
1.Write at least 500 words discussing the benefits of using R with H.docx
1.Write at least 500 words discussing the benefits of using R with H.docx1.Write at least 500 words discussing the benefits of using R with H.docx
1.Write at least 500 words discussing the benefits of using R with H.docxcroysierkathey
 
1.What is Starbucks’ ROA for 2012, 2011, and 2010 Why might focusin.docx
1.What is Starbucks’ ROA for 2012, 2011, and 2010 Why might focusin.docx1.What is Starbucks’ ROA for 2012, 2011, and 2010 Why might focusin.docx
1.What is Starbucks’ ROA for 2012, 2011, and 2010 Why might focusin.docxcroysierkathey
 
1.  Discuss the cultural development of the Japanese and the Jewis.docx
1.  Discuss the cultural development of the Japanese and the Jewis.docx1.  Discuss the cultural development of the Japanese and the Jewis.docx
1.  Discuss the cultural development of the Japanese and the Jewis.docxcroysierkathey
 
1.  Discuss at least 2  contextual factors(family, peers,  school,.docx
1.  Discuss at least 2  contextual factors(family, peers,  school,.docx1.  Discuss at least 2  contextual factors(family, peers,  school,.docx
1.  Discuss at least 2  contextual factors(family, peers,  school,.docxcroysierkathey
 
1.Write at least 500 words in APA format discussing how to use senti.docx
1.Write at least 500 words in APA format discussing how to use senti.docx1.Write at least 500 words in APA format discussing how to use senti.docx
1.Write at least 500 words in APA format discussing how to use senti.docxcroysierkathey
 
1.The following clause was added to the Food and Drug Actthe S.docx
1.The following clause was added to the Food and Drug Actthe S.docx1.The following clause was added to the Food and Drug Actthe S.docx
1.The following clause was added to the Food and Drug Actthe S.docxcroysierkathey
 
1.What are social determinants of health  Explain how social determ.docx
1.What are social determinants of health  Explain how social determ.docx1.What are social determinants of health  Explain how social determ.docx
1.What are social determinants of health  Explain how social determ.docxcroysierkathey
 
1.This week, we’ve been introduced to the humanities and have ta.docx
1.This week, we’ve been introduced to the humanities and have ta.docx1.This week, we’ve been introduced to the humanities and have ta.docx
1.This week, we’ve been introduced to the humanities and have ta.docxcroysierkathey
 
1.What are barriers to listening2.Communicators identif.docx
1.What are barriers to listening2.Communicators identif.docx1.What are barriers to listening2.Communicators identif.docx
1.What are barriers to listening2.Communicators identif.docxcroysierkathey
 
1.Timeline description and details There are multiple way.docx
1.Timeline description and details There are multiple way.docx1.Timeline description and details There are multiple way.docx
1.Timeline description and details There are multiple way.docxcroysierkathey
 
1.The PresidentArticle II of the Constitution establishe.docx
1.The PresidentArticle II of the Constitution establishe.docx1.The PresidentArticle II of the Constitution establishe.docx
1.The PresidentArticle II of the Constitution establishe.docxcroysierkathey
 
1.What other potential root causes might influence patient fal.docx
1.What other potential root causes might influence patient fal.docx1.What other potential root causes might influence patient fal.docx
1.What other potential root causes might influence patient fal.docxcroysierkathey
 

Plus de croysierkathey (20)

1.  Discuss the organization and the family role in every one of the.docx
1.  Discuss the organization and the family role in every one of the.docx1.  Discuss the organization and the family role in every one of the.docx
1.  Discuss the organization and the family role in every one of the.docx
 
1.  Compare and contrast DEmilios Capitalism and Gay Identity .docx
1.  Compare and contrast DEmilios Capitalism and Gay Identity .docx1.  Compare and contrast DEmilios Capitalism and Gay Identity .docx
1.  Compare and contrast DEmilios Capitalism and Gay Identity .docx
 
1.Purpose the purpose of this essay is to spread awareness .docx
1.Purpose the purpose of this essay is to spread awareness .docx1.Purpose the purpose of this essay is to spread awareness .docx
1.Purpose the purpose of this essay is to spread awareness .docx
 
1.  Tell us why it is your favorite film.2.  Talk about the .docx
1.  Tell us why it is your favorite film.2.  Talk about the .docx1.  Tell us why it is your favorite film.2.  Talk about the .docx
1.  Tell us why it is your favorite film.2.  Talk about the .docx
 
1.What are the main issues facing Fargo and Town Manager Susan.docx
1.What are the main issues facing Fargo and Town Manager Susan.docx1.What are the main issues facing Fargo and Town Manager Susan.docx
1.What are the main issues facing Fargo and Town Manager Susan.docx
 
1.Writing Practice in Reading a PhotographAttached Files.docx
1.Writing Practice in Reading a PhotographAttached Files.docx1.Writing Practice in Reading a PhotographAttached Files.docx
1.Writing Practice in Reading a PhotographAttached Files.docx
 
1.Some say that analytics in general dehumanize managerial activitie.docx
1.Some say that analytics in general dehumanize managerial activitie.docx1.Some say that analytics in general dehumanize managerial activitie.docx
1.Some say that analytics in general dehumanize managerial activitie.docx
 
1.What is the psychological term for the symptoms James experiences .docx
1.What is the psychological term for the symptoms James experiences .docx1.What is the psychological term for the symptoms James experiences .docx
1.What is the psychological term for the symptoms James experiences .docx
 
1.Write at least 500 words discussing the benefits of using R with H.docx
1.Write at least 500 words discussing the benefits of using R with H.docx1.Write at least 500 words discussing the benefits of using R with H.docx
1.Write at least 500 words discussing the benefits of using R with H.docx
 
1.What is Starbucks’ ROA for 2012, 2011, and 2010 Why might focusin.docx
1.What is Starbucks’ ROA for 2012, 2011, and 2010 Why might focusin.docx1.What is Starbucks’ ROA for 2012, 2011, and 2010 Why might focusin.docx
1.What is Starbucks’ ROA for 2012, 2011, and 2010 Why might focusin.docx
 
1.  Discuss the cultural development of the Japanese and the Jewis.docx
1.  Discuss the cultural development of the Japanese and the Jewis.docx1.  Discuss the cultural development of the Japanese and the Jewis.docx
1.  Discuss the cultural development of the Japanese and the Jewis.docx
 
1.  Discuss at least 2  contextual factors(family, peers,  school,.docx
1.  Discuss at least 2  contextual factors(family, peers,  school,.docx1.  Discuss at least 2  contextual factors(family, peers,  school,.docx
1.  Discuss at least 2  contextual factors(family, peers,  school,.docx
 
1.Write at least 500 words in APA format discussing how to use senti.docx
1.Write at least 500 words in APA format discussing how to use senti.docx1.Write at least 500 words in APA format discussing how to use senti.docx
1.Write at least 500 words in APA format discussing how to use senti.docx
 
1.The following clause was added to the Food and Drug Actthe S.docx
1.The following clause was added to the Food and Drug Actthe S.docx1.The following clause was added to the Food and Drug Actthe S.docx
1.The following clause was added to the Food and Drug Actthe S.docx
 
1.What are social determinants of health  Explain how social determ.docx
1.What are social determinants of health  Explain how social determ.docx1.What are social determinants of health  Explain how social determ.docx
1.What are social determinants of health  Explain how social determ.docx
 
1.This week, we’ve been introduced to the humanities and have ta.docx
1.This week, we’ve been introduced to the humanities and have ta.docx1.This week, we’ve been introduced to the humanities and have ta.docx
1.This week, we’ve been introduced to the humanities and have ta.docx
 
1.What are barriers to listening2.Communicators identif.docx
1.What are barriers to listening2.Communicators identif.docx1.What are barriers to listening2.Communicators identif.docx
1.What are barriers to listening2.Communicators identif.docx
 
1.Timeline description and details There are multiple way.docx
1.Timeline description and details There are multiple way.docx1.Timeline description and details There are multiple way.docx
1.Timeline description and details There are multiple way.docx
 
1.The PresidentArticle II of the Constitution establishe.docx
1.The PresidentArticle II of the Constitution establishe.docx1.The PresidentArticle II of the Constitution establishe.docx
1.The PresidentArticle II of the Constitution establishe.docx
 
1.What other potential root causes might influence patient fal.docx
1.What other potential root causes might influence patient fal.docx1.What other potential root causes might influence patient fal.docx
1.What other potential root causes might influence patient fal.docx
 

Dernier

ARTERIAL BLOOD GAS ANALYSIS........pptx
ARTERIAL BLOOD  GAS ANALYSIS........pptxARTERIAL BLOOD  GAS ANALYSIS........pptx
ARTERIAL BLOOD GAS ANALYSIS........pptxAneriPatwari
 
ICS 2208 Lecture Slide Notes for Topic 6
ICS 2208 Lecture Slide Notes for Topic 6ICS 2208 Lecture Slide Notes for Topic 6
ICS 2208 Lecture Slide Notes for Topic 6Vanessa Camilleri
 
Grade 9 Quarter 4 Dll Grade 9 Quarter 4 DLL.pdf
Grade 9 Quarter 4 Dll Grade 9 Quarter 4 DLL.pdfGrade 9 Quarter 4 Dll Grade 9 Quarter 4 DLL.pdf
Grade 9 Quarter 4 Dll Grade 9 Quarter 4 DLL.pdfJemuel Francisco
 
4.11.24 Poverty and Inequality in America.pptx
4.11.24 Poverty and Inequality in America.pptx4.11.24 Poverty and Inequality in America.pptx
4.11.24 Poverty and Inequality in America.pptxmary850239
 
CLASSIFICATION OF ANTI - CANCER DRUGS.pptx
CLASSIFICATION OF ANTI - CANCER DRUGS.pptxCLASSIFICATION OF ANTI - CANCER DRUGS.pptx
CLASSIFICATION OF ANTI - CANCER DRUGS.pptxAnupam32727
 
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptx
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptxBIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptx
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptxSayali Powar
 
MS4 level being good citizen -imperative- (1) (1).pdf
MS4 level   being good citizen -imperative- (1) (1).pdfMS4 level   being good citizen -imperative- (1) (1).pdf
MS4 level being good citizen -imperative- (1) (1).pdfMr Bounab Samir
 
Expanded definition: technical and operational
Expanded definition: technical and operationalExpanded definition: technical and operational
Expanded definition: technical and operationalssuser3e220a
 
How to Manage Buy 3 Get 1 Free in Odoo 17
How to Manage Buy 3 Get 1 Free in Odoo 17How to Manage Buy 3 Get 1 Free in Odoo 17
How to Manage Buy 3 Get 1 Free in Odoo 17Celine George
 
Active Learning Strategies (in short ALS).pdf
Active Learning Strategies (in short ALS).pdfActive Learning Strategies (in short ALS).pdf
Active Learning Strategies (in short ALS).pdfPatidar M
 
Indexing Structures in Database Management system.pdf
Indexing Structures in Database Management system.pdfIndexing Structures in Database Management system.pdf
Indexing Structures in Database Management system.pdfChristalin Nelson
 
4.11.24 Mass Incarceration and the New Jim Crow.pptx
4.11.24 Mass Incarceration and the New Jim Crow.pptx4.11.24 Mass Incarceration and the New Jim Crow.pptx
4.11.24 Mass Incarceration and the New Jim Crow.pptxmary850239
 
4.16.24 Poverty and Precarity--Desmond.pptx
4.16.24 Poverty and Precarity--Desmond.pptx4.16.24 Poverty and Precarity--Desmond.pptx
4.16.24 Poverty and Precarity--Desmond.pptxmary850239
 
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITW
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITWQ-Factor HISPOL Quiz-6th April 2024, Quiz Club NITW
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITWQuiz Club NITW
 
ESP 4-EDITED.pdfmmcncncncmcmmnmnmncnmncmnnjvnnv
ESP 4-EDITED.pdfmmcncncncmcmmnmnmncnmncmnnjvnnvESP 4-EDITED.pdfmmcncncncmcmmnmnmncnmncmnnjvnnv
ESP 4-EDITED.pdfmmcncncncmcmmnmnmncnmncmnnjvnnvRicaMaeCastro1
 
Tree View Decoration Attribute in the Odoo 17
Tree View Decoration Attribute in the Odoo 17Tree View Decoration Attribute in the Odoo 17
Tree View Decoration Attribute in the Odoo 17Celine George
 
4.9.24 School Desegregation in Boston.pptx
4.9.24 School Desegregation in Boston.pptx4.9.24 School Desegregation in Boston.pptx
4.9.24 School Desegregation in Boston.pptxmary850239
 
Grade Three -ELLNA-REVIEWER-ENGLISH.pptx
Grade Three -ELLNA-REVIEWER-ENGLISH.pptxGrade Three -ELLNA-REVIEWER-ENGLISH.pptx
Grade Three -ELLNA-REVIEWER-ENGLISH.pptxkarenfajardo43
 
CHEST Proprioceptive neuromuscular facilitation.pptx
CHEST Proprioceptive neuromuscular facilitation.pptxCHEST Proprioceptive neuromuscular facilitation.pptx
CHEST Proprioceptive neuromuscular facilitation.pptxAneriPatwari
 

Dernier (20)

ARTERIAL BLOOD GAS ANALYSIS........pptx
ARTERIAL BLOOD  GAS ANALYSIS........pptxARTERIAL BLOOD  GAS ANALYSIS........pptx
ARTERIAL BLOOD GAS ANALYSIS........pptx
 
ICS 2208 Lecture Slide Notes for Topic 6
ICS 2208 Lecture Slide Notes for Topic 6ICS 2208 Lecture Slide Notes for Topic 6
ICS 2208 Lecture Slide Notes for Topic 6
 
Grade 9 Quarter 4 Dll Grade 9 Quarter 4 DLL.pdf
Grade 9 Quarter 4 Dll Grade 9 Quarter 4 DLL.pdfGrade 9 Quarter 4 Dll Grade 9 Quarter 4 DLL.pdf
Grade 9 Quarter 4 Dll Grade 9 Quarter 4 DLL.pdf
 
4.11.24 Poverty and Inequality in America.pptx
4.11.24 Poverty and Inequality in America.pptx4.11.24 Poverty and Inequality in America.pptx
4.11.24 Poverty and Inequality in America.pptx
 
CLASSIFICATION OF ANTI - CANCER DRUGS.pptx
CLASSIFICATION OF ANTI - CANCER DRUGS.pptxCLASSIFICATION OF ANTI - CANCER DRUGS.pptx
CLASSIFICATION OF ANTI - CANCER DRUGS.pptx
 
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptx
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptxBIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptx
BIOCHEMISTRY-CARBOHYDRATE METABOLISM CHAPTER 2.pptx
 
MS4 level being good citizen -imperative- (1) (1).pdf
MS4 level   being good citizen -imperative- (1) (1).pdfMS4 level   being good citizen -imperative- (1) (1).pdf
MS4 level being good citizen -imperative- (1) (1).pdf
 
Expanded definition: technical and operational
Expanded definition: technical and operationalExpanded definition: technical and operational
Expanded definition: technical and operational
 
prashanth updated resume 2024 for Teaching Profession
prashanth updated resume 2024 for Teaching Professionprashanth updated resume 2024 for Teaching Profession
prashanth updated resume 2024 for Teaching Profession
 
How to Manage Buy 3 Get 1 Free in Odoo 17
How to Manage Buy 3 Get 1 Free in Odoo 17How to Manage Buy 3 Get 1 Free in Odoo 17
How to Manage Buy 3 Get 1 Free in Odoo 17
 
Active Learning Strategies (in short ALS).pdf
Active Learning Strategies (in short ALS).pdfActive Learning Strategies (in short ALS).pdf
Active Learning Strategies (in short ALS).pdf
 
Indexing Structures in Database Management system.pdf
Indexing Structures in Database Management system.pdfIndexing Structures in Database Management system.pdf
Indexing Structures in Database Management system.pdf
 
4.11.24 Mass Incarceration and the New Jim Crow.pptx
4.11.24 Mass Incarceration and the New Jim Crow.pptx4.11.24 Mass Incarceration and the New Jim Crow.pptx
4.11.24 Mass Incarceration and the New Jim Crow.pptx
 
4.16.24 Poverty and Precarity--Desmond.pptx
4.16.24 Poverty and Precarity--Desmond.pptx4.16.24 Poverty and Precarity--Desmond.pptx
4.16.24 Poverty and Precarity--Desmond.pptx
 
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITW
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITWQ-Factor HISPOL Quiz-6th April 2024, Quiz Club NITW
Q-Factor HISPOL Quiz-6th April 2024, Quiz Club NITW
 
ESP 4-EDITED.pdfmmcncncncmcmmnmnmncnmncmnnjvnnv
ESP 4-EDITED.pdfmmcncncncmcmmnmnmncnmncmnnjvnnvESP 4-EDITED.pdfmmcncncncmcmmnmnmncnmncmnnjvnnv
ESP 4-EDITED.pdfmmcncncncmcmmnmnmncnmncmnnjvnnv
 
Tree View Decoration Attribute in the Odoo 17
Tree View Decoration Attribute in the Odoo 17Tree View Decoration Attribute in the Odoo 17
Tree View Decoration Attribute in the Odoo 17
 
4.9.24 School Desegregation in Boston.pptx
4.9.24 School Desegregation in Boston.pptx4.9.24 School Desegregation in Boston.pptx
4.9.24 School Desegregation in Boston.pptx
 
Grade Three -ELLNA-REVIEWER-ENGLISH.pptx
Grade Three -ELLNA-REVIEWER-ENGLISH.pptxGrade Three -ELLNA-REVIEWER-ENGLISH.pptx
Grade Three -ELLNA-REVIEWER-ENGLISH.pptx
 
CHEST Proprioceptive neuromuscular facilitation.pptx
CHEST Proprioceptive neuromuscular facilitation.pptxCHEST Proprioceptive neuromuscular facilitation.pptx
CHEST Proprioceptive neuromuscular facilitation.pptx
 

linical pharmacogenomics consists of the appli-cation of res.docx

  • 1. linical pharmacogenomics consists of the appli- cation of research that links measurable genetic variants with the prediction of drug response.1 Every medical specialty can utilize the results of pharmacogenomic probe studies to inform the adoption of individualized pharmacotherapy. However, psychiatric pharmacother- apy is particularly likely to benefit from the introduction of pharmacogenomic testing, because there are many psychotropic agents available for selection that target specific symptoms. The terms pharmacogenetics and pharmacogenomics are currently used interchangeably. However, with the growing understanding that multiple intragenic varia- tions should be considered in making predictions related to medication response, the use of the term pharma- cogenomics has become more frequently chosen to des- ignate the process of using documented genetic variation to guide medication selection and dosing. Historically, psychiatrists have used empirical strategies to select medications. In the best practices, the choice of medications has evolved based on a rational trial-and- error process that has used clinical indicators to select medications and then relied on documenting treatment responses to titrate the optimal dose for a particular patient. Psychiatrists learn to “start low and go slow” in order to minimize side effects. They also know that it is 6 9 P h a r m a c o l o g i c a l a s p e c t s C
  • 2. Copyright © 2010 LLS SAS. All rights reserved www.dialogues-cns.org Psychiatric pharmacogenomic testing in clinical practice David A. Mrazek, MD, FRCPsych Keywords: pharmacogenomic testing; cytochrome P450 gene; serotonin trans- porter gene (SLC6A4); serotonin receptor 2A gene (HTR2A); serotonin receptor 2C gene (HTR2C); poor metabolizer phenotype; ultrarapid metabolizer phenotype Author affiliations: Chair, Department of Psychiatry and Psychology, Mayo Clinic; Professor of Psychiatry and Pediatrics, Mayo Clinic College of Medicine, Rochester, Minnesota, USA Address for correspondence: David A. Mrazek, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA (e-mail: [email protected]) The clinical adoption of psychiatric pharmacogenomic testing has taken place rapidly over the past 7 years. Initially, drug-metabolizing enzyme genes, such as the cytochrome P450 2D6 gene (CYP2D6), were identified. Genotyping the highly variable cytochrome P450 2D6 gene now provides clinicians with the opportunity to iden- tify both poor metabolizers and ultrarapid metabolizers of 2D6 substrate medications. Subsequently, genes influ- encing the pharmacodynamic response of medications have been made available for clinical practice. Among the
  • 3. earliest “target genes” was the serotonin transporter gene (SLC6A4) which has variants that have been shown to influence the clinical response of patients of European ancestry when they are treated with selective serotonin reuptake inhibitors. Genotyping of some of the serotonin receptor genes is also available to guide clinical practice. The quantification of the clinical utility of pharmacoge- nomic testing is evolving, and ethical considerations for testing have been established. Given the increasingly clear cost-effectiveness of genotyping, it has recently been pre- dicted that pharmacogenomic testing will routinely be ordered to guide the selection and dosing of psychotropic medications. © 2010, LLS SAS Dialogues Clin Neurosci. 2010;12:69-76. DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 69 necessary to provide their patients with an “adequate” trial of each medication. Unfortunately, these strategies can result in a 3- to 4-week interval during which the patient continues to experience symptoms. In recent years, the potential iatrogenic harm associated with psy- chotropic medications has become increasingly obvious, with “black-box warnings” being attached to antide- pressants, antipsychotic medications, stimulants, and mood stabilizers. Despite a growing awareness of this potential harm, there are powerful pressures to try to accelerate the achievement of therapeutic benefit. At the most basic level, patients are impatient. They do not want to wait a month to achieve symptom relief. Additionally, with an increasing focus on the relief of specific symptoms, strategies using multiple psychotropic medications have become a standard of practice. Research supports the
  • 4. common practice of augmenting an initial medication with a second psychotropic drug.2 However, there is no scientifically available evidence to support the practice of using four or five psychotropic medications simulta- neously. Nevertheless, patients routinely receive multi- ple psychotropic medications in an attempt to identify the “right combination.” While some patients do achieve a good therapeutic response using this trial-and-error approach to individualized medicine, it is also true that others become overmedicated or suffer from iatrogenic side effects. Pharmacogenomic testing provides an innovative strat- egy to improve the likelihood of selecting an effective psychotropic medication. The earliest medical texts rec- ognize that individual patients experience quite dra- matically different responses to the same drug. There is also a longstanding observation that unusual drug responses can occur in members of the same family. The identification of specific gene variants associated with idiosyncratic responses is about 50 years old,3 and the recognition that some psychiatric patients metabolize antidepressants at dramatically different rates has been documented for several decades.4 However, with the use of newer antidepressant medications that rarely have life-threatening complications, the relatively expensive practice of monitoring the serum levels of newer anti- depressant medications has become uncommon in the United States. This change has occurred despite the fact that serum levels of these newer agents also have dra- matic variations based on the metabolic capacity of each patient. A decade ago, the cost of genotyping began to become more affordable, and individual laboratories initiated pharmacogenomic testing that would provide genotyping of individual cytochrome P450 genes. However, there was
  • 5. no standard or well-validated methodology for the geno- typing of these informative genes. There was also consid- erable variability in the interpretation of the results. In 2004, the US Food and Drug Administration (FDA) approved the use of a new product, the AmpliChip.5 The introduction of the AmpliChip provided reference labo- ratories with a standard method for identifying variations in two of the cytochrome P450 genes: cytochrome P450 2D6 (CYP2D6) and cytochrome P450 2C19 (CYP2C19). The approval of the AmpliChip was an important land- mark in the history of psychiatric pharmacogenomic test- ing, and within 3 years, CYP2D6 and CYP2C19 were being genotyped by every reference laboratory in the country. However, this advance also highlighted some of the challenges associated with the introduction of clinical testing. One of the most obvious challenges that must be addressed is how to begin to assess new variants of these two genes in updated versions of the assay. Ideally, the methodology for establishing drug-metabolizing pheno- types should be updated regularly based on new molecu- lar genetic findings showing how new genotypic variants influence gene function. Also, the clarification of the pre- dictive capacity of previously identified gene variants influencing gene function is similarly evolving, and newly identified associations between gene structure and func- tion should ideally be incorporated into algorithms that define the metabolic capacity of psychiatric patients. The evolution of pharmacogenomic research should inform modifications in pharmacogenomic testing. However, an implication of the rapid increase in our knowledge base is that these new studies demonstrate limitations in the accuracy of older genotyping method- ologies that were designed prior to the discovery of more recent variants. What is often not well appreciated is that even older pharmacogenomic methods provided important information for many patients, as these early
  • 6. innovations were a major advance over psychopharma- cological practice without pharmacogenomic insights. However, as newer methodologies have further improved the accuracy of the prediction of medication response, the clinical utility of pharmacogenomic testing continues to increase. Pharmacogenomic testing in psychiatric practice initially focused on identifying pharmacokinetic variability that P h a r m a c o l o g i c a l a s p e c t s 7 0 DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 70 would influence the responses of patients who had atyp- ical genotypes. Pharmacokinetic variation influences the concentration of a drug at its sites of action. Pharmacogenomic testing of drug-metabolizing enzyme genes provides a prediction of how an individual patient will metabolize a specific psychotropic medication. More recently, the focus of pharmacogenomic testing has expanded to include determining variability in the phar- macodynamic response of a patient to a specific med- ication. This variability reflects the capacity of the indi- vidual patient to respond to adequate exposure to the drug. Prediction of response is estimated based on the documentation of variations in “target genes” that code for receptors and transporters that influence the response of the patient to a particular medication. This review will first identify the most widely genotyped drug-metabolizing enzyme genes that influence the pharmacokinetic metabolic capacity of a patient. Then, it will focus on genes that influence the pharmacody-
  • 7. namic responses of individual patients, before conclud- ing with a brief discussion of the clinical utility of phar- macogenomic testing and some of the ethical considerations related to its routine use. Pharmacogenomic testing to establish the metabolic capacity of psychiatric patients Many genes code for enzymes that influence drug response. However, only the clinical implications of geno- typing four of the most commonly tested cytochrome P450 genes will be reviewed. The focus of this discussion will be the clinical benefit for the patient of identifying individualized molecular variations, and the implications for those patients who have a quite significant decrement in their capacity to metabolize specific psychotropic med- ications. Identifying these individual patients provides clinicians with a clear method of minimizing side effects. This determination of decreased metabolic capacity is the most obvious benefit of pharmacogenomic testing, but implications of the pharmacogenomic testing for patients with increased metabolic capacity will also be discussed, as these patients are less likely to respond to specific psy- chotropic medications. The cytochrome P450 2D6 gene (CYP2D6) CYP2D6 was the first drug-metabolizing enzyme gene that was genotyped to identify psychiatric patients with increased or decreased metabolic capacity. It is located on chromosome 22 and consists of 4382 nucleotides. CYP2D6 codes for an enzyme that is composed of 497 amino acids. The CYP2D6 enzyme plays a primary role in the metab- olism of more than 70 substrate medications, including
  • 8. twelve psychotropic medications. CYP2D6 is one of the most highly variable drug-metabolizing enzyme genes. However, many of the other 29 P450 drug-metabolizing enzyme genes are also highly variable. The specific genetic variations that define variable phenotypes can be located on a Web site maintained by the Karolinska Institute (http://www.cypalleles.ki.se/). Each newly iden- tified variant is included on the Web site after confir- mation that it is unique. There are currently 75 distinct CYP2D6 alleles posted on this site, as well as an additional 55 CYP2D6 variants that closely resemble one of the primary variants. Traditionally, these variants have been classified as being normal, deficient, or inactive drug-metabolizing alleles. Additionally, some alleles have more recently been demonstrated to code for an increased amount of enzyme which enhances the metabolic activity of the patient. Furthermore, patients can have a variable num- ber of copies of CYP2D6. The most common number of copies of CYP2D6 that patients carry is two. However, some patients have only one copy and, rarely, none at all. It is also possible to have more than two copies, and one patient has been reported to have 13 copies.4 The devel- opment of several different classification systems to cat- egorize 2D6 substrate metabolic capacity of patients into four phenotypic categories has been problematic. The use of alternative methodologies by different research teams has made it more difficult to study the implica- tions of this variability. The most important CYP2D6 phenotype to identify is the poor 2D6 substrate metabolizer phenotype. Patients who are poor metabolizers are at increased risk for adverse events when they are prescribed 2D6 substrate medications, because of their low metabolic capacity. Patients are now classified as poor metabolizers if they have two inactive alleles, or one inactive allele and one
  • 9. deficient allele. The second most clinically important CYP2D6 pheno- type is the ultrarapid metabolizer phenotype. Patients are ultrarapid metabolizers if they have either three or more active copies of CYP2D6 or two or more enhanced copies of CYP2D6. They are unlikely to respond to 2D6 Psychiatric pharmacogenomic testing - Mrazek Dialogues in Clinical Neuroscience - Vol 12 . No. 1 . 2010 7 1 DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 71 substrate medications at standard doses because their ability to rapidly metabolize these medications makes it difficult to sustain therapeutic serum levels. The third clinically important CYP2D6 phenotype is the intermediate metabolizer phenotype. These patients have one normal copy of CYP2D6, and one copy that is either deficient or inactive. While these patients can nor- mally benefit from 2D6 substrate medications at low-to- moderate doses, they are at increased risk for the devel- opment of side effects at higher doses because of their decreased metabolic capacity, and they are more at risk for enzyme inhibition as a consequence of drug-drug interactions. When intermediate metabolizers are exposed to powerful 2D6 inhibitors such as paroxetine or fluoxetine, their metabolic capacity can be further decreased to the level of a poor metabolizer.6 There are many psychotropic medications metabolized by the 2D6 enzyme. Specifically, this enzyme: • primarily metabolizes five antidepressants: fluoxetine,
  • 10. paroxetine, venlafaxine, desipramine, and nortriptyline • substantially metabolizes amitriptyline, imipramine, doxepin, duloxetine, trazodone, and mirtazapine • primarily metabolizes risperidone and four of the typ- ical antipsychotic medications: chlorpromazine, thior- idazine, perphenazine, and haloperidol • has substantial involvement in the metabolism of arip- iprazole and olanzapine • primarily metabolizes atomoxetine and dextroam- phetamine. Beyond the prescription of psychotropic medications, psychiatric patients are given many other 2D6 substrate medications. Specifically, dextromethorphan is a cough suppressant that is metabolized by the 2D6 enzyme. Patients who are poor metabolizers of 2D6 substrate medications are at increased risk for cognitive side effects if taking standard doses of preparations that contain dex- tromethorphan. Another example is codeine, which is a prodrug. A prodrug must be converted to an active metabolite in order to have a therapeutic effect. Patients who are poor 2D6 metabolizers do not receive analgesic benefit from codeine because they do not metabolize codeine to morphine. Tamoxifen is also a prodrug that is the most frequently prescribed treatment for breast can- cer. Poor metabolizers have little or no benefit from tamoxifen because they are not able to metabolize tamoxifen to endoxifen.7,8 Additionally, paroxetine, flu- oxetine, or bupropion should not be given to patients who are receiving tamoxifen because they inhibit the
  • 11. 2D6 enzyme. Giving these inhibitors to intermediate metabolizers can convert them to functional poor metab- olizers. Consequently, they become unable to produce endoxifen.9 The cytochrome P450 2C19 gene (CYP2C19) CYP2C19 was the second drug-metabolizing enzyme gene that was widely genotyped to identify patients with increased or decreased metabolic capacity. It is a large gene located on chromosome 10. It consists of 90 209 nucleotides, but codes for an enzyme that contains only 490 amino acids. The identification of patients with low 2C19 metabolic capacity is clinically important because it allows clini- cians to decrease the risk of iatrogenic side effects. The 2C19 enzyme: • primarily metabolizes citalopram, escitalopram, clomipramine, and amitriptyline • has substantial involvement in the metabolism of ser- traline, imipramine, nortriptyline, and doxepin • plays an important role in the metabolism of clozapine and a minimal role in the metabolism of thioridazine • is the primary enzyme involved in the metabolism of diazepam. Recently, a new variant of CYP2C19 has been identified which has enhanced function.10 Patients who are homozygous for this new allele are less likely to respond to 2C19 substrate medications at standard doses. The identification of ultrarapid 2C19 metabolizers can be helpful in evaluating patients who do not respond to standard doses of any of these psychotropic medications.
  • 12. The cytochrome P450 2C9 gene (CYP2C9) CYP2C9 is located on chromosome 10 in relative close proximity to CYP2C19. However, it is only about half the size of CYP2C9 as it consists of 50 708 nucleotides. Like CYP2C19, CYP2C9 codes for an enzyme that con- tains 490 amino acids. CYP2C9 is a drug-metabolizing enzyme gene that is less routinely genotyped to identify the increased or decreased metabolic capacity of psychiatric patients for 2C9 substrate medications. It does not play a primary role in the metabolism of any currently prescribed psy- chotropic medications. However, the 2C9 enzyme pro- vides the only secondary pathway for the metabolism of fluoxetine, so patients who are poor metabolizers of P h a r m a c o l o g i c a l a s p e c t s 7 2 DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 72 Psychiatric pharmacogenomic testing - Mrazek Dialogues in Clinical Neuroscience - Vol 12 . No. 1 . 2010 7 3 both 2D6 substrates and 2C9 substrates are at very high risk for adverse effects if treated with standard dose of fluoxetine. The cytochrome P450 1A2 gene (CYP1A2)
  • 13. CYP1A2 is a less well-studied drug-metabolizing enzyme gene, but it codes for an enzyme that plays an important role in the metabolism of fluvoxamine. It is also involved in the metabolism of duloxetine and olan- zapine. CYP1A2 is located on chromosome 15 and con- sists of 7758 nucleotides. CYP1A2 codes for an enzyme that is composed of 516 amino acids. A somewhat atypical aspect of the CYP1A2 gene is that there are alleles of this gene that are inducible by smok- ing tobacco or consuming cruciferous vegetables, such as cabbage or Brussels sprouts. Consequently, patients who smoke tobacco and have two alleles of CYP1A2 that are inducible by their smoking can be difficult to maintain on 1A2 substrate medications. A relatively common problem occurs when these patients are treated with olanzapine or clozapine on an inpatient psychiatric unit that does not allow them to smoke. When they begin to smoke after they are discharged, their serum level drops and their psychotic symptoms often reoccur. In some populations of European ancestry, as many as 25% of the population can have an inducible ultrarapid CYP1A2 phenotype. Pharmacogenomic testing to identify variability in pharmacodynamic responses A goal of individualized molecular psychopharmacology is to identify medications for an individual patient that will not only be safe, but will be effective. Progress in making predictions of medication response has occurred, and while the goal of being able to predict this response with certainty has not been achieved, we can make increasingly accurate probabilistic predictions of the likelihood of response. Psychiatrists are familiar with this limitation. While hundreds of randomized clinical trials of psychotropic medications have been conducted to
  • 14. identify effective psychotropic drugs, the results of these trials only provide assurance that for a sample of patients there is reasonable likelihood that the medica- tion will be of more benefit than a placebo. While selec- tive serotonin reuptake blockers are among the most widely prescribed medications in the world, many patients do not respond. Specifically, the largest clinical effectiveness study of citalopram reported that less than 30% of the entire sample of patients experienced a com- plete remission of their symptoms.11 While the ultimate goal of pharmacodynamically designed pharmacoge- nomic testing is to identify a drug for a specific patient that will definitely be effective, at the current stage of our understanding, it is only possible to identify a med- ication that is more likely to be effective. The serotonin transporter gene (SLC6A4) SLC6A4 is located on chromosome 17 and consists of 37 800 nucleotides. It codes for an enzyme that is composed of 630 amino acids. SLC6A4 is the most widely genotyped pharmacoge- nomic “target” gene. A meta-analysis of studies of the relationship between the more active long form of the indel promoter variant of this gene and responses to selective serotonin reuptake inhibitors12 confirmed the early finding that the long form is associated with a more rapid and better response.13 However, this has not con- sistently been demonstrated in patients of Asian ances- try.14,15 The importance of ancestral heritage has been fur- ther demonstrated by multiple analyses of the large STAR*D effective treatment study. Analyses that did not consider ancestral background did not demonstrate a significant association,16 while those that focused on patients who identified themselves as
  • 15. “white” but not “Hispanic” did confirm the relationship that patients who were homozygous for the more active long form of the indel promoter polymorphism were more likely to respond to citalopram. Other variants, such as rs2553117 and the second intronic VNTR18 are likely to influence the activity level of the gene and, con- sequently, its response to medications that block its abil- ity to reuptake serotonin in the synapses of the central nervous system. The serotonin receptor 2A gene (HTR2A) HTR2A is located on chromosome 13 and consists of 62 663 nucleotides. Despite its large size, it codes for an enzyme that is composed of only 471 amino acids. There have been a series of studies examining the asso- ciation between variants of HTR2A and antidepressant response. A large study examining the response of depressed patients of European ancestry to citalopram DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 73 found that a positive response to citalopram was associ- ated with having a copy of the adenine allele of rs7997012.19 Another study examining a different HTR2A variant, rs6313, reported that patients who were homozygous for the cytosine allele were less likely to tolerate taking paroxetine than those who had one or more copies of the thymine allele.20 A series of studies have reported a better response to clozapine in patients who had the thymine allele of
  • 16. rs6313. The thymine allele of rs6313 has also been asso- ciated with a lower risk for the development of extrapyramidal side effects when taking antipsychotic medications.21-23 The serotonin receptor 2C gene (HTR2C) HTR2C is a very large gene that is located on the X chromosome and consists of 326 074 nucleotides. However, it codes for a protein product that is composed of only 458 amino acids. Variations in the HTR2C gene have been associated with a better clinical response to clozapine. Specifically, patients with schizophrenia who have a copy of the cyto- sine allele of rs6318 have achieved better control of their psychotic symptoms than patients with the guanine allele.24,25 However, this same variant has been associated with a higher risk for the development of extrapyrami- dal side effects in patients who are taking typical antipsy- chotic medications.26 An increased risk for the development of weight gain has been linked to a different HTR2C variant. Specifically, the cytosine allele of rs518147 is associated with increased weight gain, while the thymine allele is conceptualized as providing protection against weight gain.27-29 The clinical utility of pharmacogenomic testing in psychiatric practice Assessing the clinical utility of pharmacogenomic test- ing is an ongoing process, given that the accuracy of genotyping is continually improving, and new research is identifying additional genetic variants that influence medication responses. Reports of adverse responses to
  • 17. 2D6 substrate medications in patients with decreased 2D6 metabolic capacity support the use of testing at this most basic level. Specifically, poor 2D6 metabolizers have had quite dramatic side effects to 2D6 substrate medications3 and some toxic reactions have been lethal.30,31 However, there have been no large random- ized clinical trials to demonstrate the clinical utility of pharmacogenomic testing. Such trials would reinforce the use of testing. However, it is unlikely that these tri- als will ever be conducted because, by definition, they are not designed to concentrate on those patients who are the most likely to benefit from pharmacogenomic testing. Trials that screen vulnerable populations and identify patients at risk for suboptimal responses to med- ications are a more efficient method to address the clin- ical usefulness of testing patients with decreased meta- bolic capacity. These screened patients could then be enrolled in protocols designed to provide optimal response for their specific genotypes and predicted phar- macogenomic phenotypes. Ethical considerations for pharmacogenomic testing in psychiatric practice The provision of pharmacogenomic testing involves rel- atively few risks, but ethical safeguards are still impor- tant to consider. These are essentially the same consid- erations that are important to think through when ordering any laboratory test that has the potential to direct a treatment decision. First, clinical pharmacogenomic testing requires obtain- ing appropriate consent. This has become a guiding prin- ciple for all diagnostic and therapeutic procedures. Clinicians should provide the basic rationale for pro- ceeding with pharmacogenomic testing so that their
  • 18. patients have the opportunity to provide explicit informed consent. Secondly, as a component of obtaining clinical consent, it must be clear that clinical testing is a voluntary proce- dure. This is true for virtually all clinical laboratory test- ing with the relatively rare exceptions of mandatory test- ing that can identify a condition with a potential negative influence on the public health of the commu- nity. A common example of compulsory testing is the monitoring of infections in order to prevent contagion. A third principle is that clinicians must insure the confi- dentiality of sensitive medical information that becomes a part of the medical record of the patient. This is true whether the information is derived from a pathological specimen that reveals a malignant carcinoma or from magnetic resonance imaging that demonstrates atrophy P h a r m a c o l o g i c a l a s p e c t s 7 4 DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 74 of the hippocampus. The security of the medical record is the responsibility of the clinician. Finally, any diagnostic medical procedure must have an acceptable level of reliability. The degree of accuracy of any clinical laboratory testing is dependent on a number of key variables. Two of these variables are the serious- ness of the prognosis for the patient if the test is positive and the efficacy of available treatments. In designing the treatment plan for a potentially lethal condition that is likely to respond well to a relatively benign intervention if it is administered early in the course of the illness, a
  • 19. laboratory test with high sensitivity is desirable. The most important objective in this situation is to identify as quickly as possible those patients who will benefit from treatment. Future developments that will influence pharmacogenomic testing in psychiatric practice In the 2009 presidential lecture of the American Psychiatric Association, it was predicted that pharma- cogenomic testing would become a part of everyday psy- chiatric practice.32 Ironically, in many academic health centers, pharmacogenomic testing has been utilized since 2004—the time of the introduction of the AmpliChip. Over the intervening years, early adopters have inte- grated pharmacogenomic testing into their inpatient protocols and ultimately into their outpatient practices. However, this testing has not yet been included in many clinical guidelines. Pharmacogenomic testing is an innovation, and it takes time for innovations to become integrated into standard practice. While it is difficult to predict with accuracy just how quickly pharmacogenomic testing will become an essential component of clinical psychopharmacological practice, there is no question that this will happen. Ironically, given advances in our ability to sequence genes both rapidly and inexpensively, there will come a time in the near future when most patients will know their 2D6 phenotype in the same way as today they know their blood type. However, well before we reach a state of uni- versal awareness of our informative genotypes, our patients will no longer accept avoidable side effects, and will demand basic pharmacogenomic testing prior to tak-
  • 20. ing antidepressant or antipsychotic medications. ❏ Psychiatric pharmacogenomic testing - Mrazek Dialogues in Clinical Neuroscience - Vol 12 . No. 1 . 2010 7 5 REFERENCES 1. Mrazek DA. Psychiatric Pharmacogenomics. New York, NY: Oxford University Press; 2010. 2. Thase ME, Friedman ES, Biggs MM, et al. Cognitive therapy versus med- ication in augmentation and switch strategies as second-step treatments: a STAR*D report. Am J Psychiatry. 2007;164:739-752. 3. Weinshilboum R. Inheritance and drug response. N Engl J Med. 2003;348:529-537. 4. Dalen P, Dahl ML, Ruiz ML, Nordin J, Bertilsson L. 10- Hydroxylation of nortriptyline in white persons with 0, 1, 2, 3, and 13 functional CYP2D6 genes. Clin Pharmacol Ther. 1998;63:444-452. 5. Mrazek D. Out of the pipeline: pharmacogenomic DNA chip. Curr Psychiatr. 2005;4:67-73. 6. Preskorn S. Reproducibility of the in vivo effect of the selective sero- tonin reuptake inhibitors on the in vivo function of cytochrome P450 2D6: an update (part II). J Psychiatr Pract. 2003;9:150-158. 7. Goetz MP, Knox SK, Suman VJ, et al. The impact of cytochrome P450 2D6 metabolism in women receiving adjuvant tamoxifen. Breast
  • 21. Cancer Res Treat. 2007;101:113-21. 8. Schroth W, Goetz MP, Hamann U, et al. Associaiton between CYP2D6 polymorphisms and outcomes among women with early stage breast can- cer treated with tamoxifen. JAMA. 2009;302:1429-1436. 9. Henry NL, Stearns V, Flockhart DA, Hayes DF, Riba M. Drug interactions and pharmacogenomics in the treatment of breast cancer and depression. Am J Psychiatry. 2008;165:1251-1255. 10. Sim SC, Risinger C, Dahl ML, et al. A common novel CYP2C19 gene vari- ant causes ultrarapid drug metabolism relevant for the drug response to proton pump inhibitors and antidepressants. Clin Pharmacol Ther. 2006;79:103-113. 11. Trivedi M, Rush A, Wisniewski S, et al. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: impli- cations for clinical practice. Am J Psychiatry. 2006;163:28. 12. Serretti A, Kato M, De Ronchi D, Kinoshita T. Meta- analysis of serotonin transporter gene promoter polymorphism (5-HTTLPR) association with selec- tive serotonin reuptake inhibitor efficacy in depressed patients. Mol Psychiatry. 2007;12:247-257. 13. Serretti A, Zanardi R, Rossini D, Cusin C, Lilli R, Smeraldi E. Influence of tryptophan hydroxylase and serotonin transporter genes on fluvoxamine
  • 22. antidepressant activity. Mol Psychiatry. 2001;6:586-592. 14. Yoshida K, Itoa K, Satoa K, et al. Influence of the serotonin transporter gene-linked polymorphic region on the antidepressant response to fluvox- amine in Japanese depressed patients. Prog Neuropsychopharmacol Biol Psychiatry. 2002;26:383-386. 15. Kim DK, Lim SW, Lee S, et al. Serotonin transporter gene polymorphism and antidepressant response. Neuroreport. 2000;11:215-219. 16. Peters E, Slager S, McGrath P, Knowles J, Hamilton S. Investigation of serotonin-related genes in antidepressant response. Mol Psychiatry. 2004;9:879-889. 17. Hu XZ, Rush AJ, Charney D, et al. Association between a functional serotonin transporter promoter polymorphism and citalopram treatment in adult outpatients with major depression. Arch Gen Psychiatry. 2007;64:783- 792. 18. Mrazek DA, Rush AJ, Biernacka JM, et al. SLC6A4 variation and citalo- pram response. Am J Med Genet Part B. 2009;150:341-351. 19. McMahon FJ, Buervenich S, Charney D, et al. Variation in the gene encoding the serotonin 2A receptor is associated with outcome of antide- pressant treatment. Am J Hum Genet. 2006;78:804-814. 20. Murphy GM, Kremer C, Rodrigues HE, Schatzberg AF. Pharmacogenetics of antidepressant medication intolerance. Am J Psychiatry. 2003;160:1830- 1835.
  • 23. DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 75 P h a r m a c o l o g i c a l a s p e c t s 7 6 Pruebas farmacogenómicas en la práctica clínica psiquiátrica La incorporación en psiquiatría clínica de las prue- bas farmacogenómicas ha ocurrido rápidamente en los últimos siete años. Inicialmente se identificaron genes de enzimas metabolizadoras de fármacos, como el gen del citocromo P450 2D6. La tipificación del gen del citocromo P450 2D6 que es altamente variable da la oportunidad actualmente a los clíni- cos de identificar a los metabolizadores pobres y los ultrarrápidos para las sustancias que son sustrato del 2D6. Con posterioridad se ha podido disponer en la práctica clínica de genes que influyen en la respuesta farmacodinámica de los medicamentos. Entre los primeros “genes blanco” estuvo el gen del transportador de serotonina (SLC6A4), el cual tiene variantes que han demostrado que influyen en la respuesta clínica de los pacientes con ancestros europeos cuando son tratados con inhibidores selectivos de la recaptura de serotonina. La tipifi- cación de algunos de los genes del receptor de sero- tonina también está disponible para guiar la prác- tica clínica. La cuantificación de la utilidad clínica de las pruebas farmacogenómicas está en desarrollo y se han establecido las consideraciones éticas para su realización. Considerando la cada vez más clara
  • 24. costo-eficacia de la tipificación génica, reciente- mente se ha pronosticado que las pruebas farma- cogenómicas se solicitarán de rutina para orientar la selección y dosificación de los fármacos psicotró- picos. Évaluation pharmacogénomique psychiatrique en pratique clinique L’évaluation pharmacogénomique psychiatrique s’est rapidement imposée en pratique clinique au cours de ces 7 dernières années. Les gènes d’en- zymes métabolisant les médicaments, comme le cytochrome P450 2D6 (CYP2D6), ont d’abord été identifiés. Le génotypage de ce gène très variable permet maintenant aux cliniciens d’identifier des métaboliseurs lents et des métaboliseurs ultrara- pides des substrats du 2D6. Des gènes influant sur la réponse pharmacodynamique des médicaments sont ensuite devenus disponibles en pratique cli- nique. Parmi les premiers « gènes cibles », le gène du transporteur de la sérotonine (SLC6A4) possède des variants qui influent sur la réponse clinique des patients d’ascendance européenne lorsqu’ils sont traités avec des inhibiteurs sélectifs de la recapture de la sérotonine. Le génotypage de certains gènes du récepteur de la sérotonine est également dis- ponible pour guider la réponse clinique. La quan- tification de l’utilité clinique de l’évaluation phar- macogénomique évolue et fait l’objet de considérations éthiques. Il a été récemment prédit qu’en raison de l’évidente rentabilité croissante du génotypage, l’évaluation pharmacogénomique devrait faire partie des examens de routine pour sélectionner et ajuster la posologie des médica- ments psychotropes.
  • 25. 21. Segman RH, Heresco-Levy U, Finkel B, et al. Association between the serotonin 2A receptor gene and tardive dyskinesia in chronic schizophre- nia. Mol Psychiatry. 2001;6:225-229. 22. Tan EC, Chong SA, Mahendran R, Dong F, Tan CH. Susceptibility to neu- roleptic-induced tardive dyskinesia and the T102C polymorphism in the sero- tonin type 2A receptor. Biol Psychiatry. 2001;50:144-147. 23. Lattuada E, Cavallaro R, Serretti A, Lorenzi C, Smeraldi E. Tardive dyski- nesia and DRD2, DRD3, DRD4, 5-HT2A variants in schizophrenia: an associa- tion study with repeated assessment. Int J Neuropsychopharmcol 2004;7:489-493. 24. Sodhi MS, Arranz MJ, Curtis D, et al. Association between clozapine response and allelic variation in the 5-HT2C receptor gene. Neuroreport. 1995;7:169-172. 25. Arranz MJ, Munro J, Birkett J, et al. Pharmacogenetic prediction of clozapine response. Lancet. 2000;355:1615-1616. 26. Gunes A, Scordo MG, Jaanson P, Dahl ML. Serotonin and dopamine receptor gene polymorphisms and the risk of extrapyramidal side effects in perphenazine-treated schizophrenic patients. Psychopharmacology 2007;190:479-484. 27. Reynolds GP, Zhang ZJ, Zhang XB. Association of antipsychotic drug- induced weight gain with a 5-HT2C receptor gene
  • 26. polymorphism. Lancet. 2002;359:2086-2087. 28. Templeman LA, Reynolds GP, Arranz B, San L. Polymorphisms of the 5- HT2C receptor and leptin genes are associated with antipsychotic drug- induced weight gain in Caucasian subjects with a first-episode psychosis. Pharmacogenet Genomics. 2005;15:195-200. 29. De Luca V, Muller DJ, Hwang R, et al. HTR2C haplotypes and antipsy- chotics-induced weight gain: X-linked multimarker analysis. Hum Psychopharmacol. 2007;22:463-467. 30. Sallee FR, DeVane CL, Ferrell RE. Fluoxetine-related death in a child with cytochrome P-450 2D6 genetic deficiency. J Child Adolesc Psychopharmacol. 2000;10:27-34. 31. Koski A, Ojanpera I, Vuori E, Sajantila A. A fatal doxepin poisoning associated with a defective CYP2D6 genotype. Am J Forensic Med Pathol. 2007;28:259-261. 32. Stotland NL. Presidential address. Am J Psychiatry. 2009;166:1100-1104. DCNS_44_5.qxd:DCNS#44 10/03/10 1:44 Page 76 E-Mail [email protected] Editorial
  • 27. Psychother Psychosom 2016;85:129–135 DOI: 10.1159/000443512 The Limitations of Genetic Testing in Psychiatry Steven L. Dubovsky Department of Psychiatry, State University of New York at Buffalo, Buffalo, N.Y. , and Departments of Psychiatry and Medicine, University of Colorado, Denver, Colo. , USA tocols for them [4] . In moderately differentiated breast cancers, which comprise 50% of breast tumors, gene ex- pression signatures for mitotic index, angiogenic poten- tial, p53 mutational status, and estrogen and progester- one dependence provide better stratification of prognosis than histology [6] . However, despite such advances, there is still not much clear integration between genomics and clinical practice in oncology [7] . Psychiatric Diagnosis Numerous markers have been associated with psychi- atric disorders, including genes for BDNF (brain-derived neurotrophic factor), FOS (FBJ murine osteosarcoma vi- ral oncogene homolog), COMT, DRD1, DRD2, DISC1, GABABR1 (γ-aminobutyric acid B receptor 1), NR4A2 (nuclear receptor subfamily 4, group A, member 2), ADORA2A (adenosine A2a receptor), CACNA1C (cal- cium channel gene), sirtuin 1, LHPP, 5HTR1A, RNA- binding proteins, and genes for myelination, glutaminer- gic and GABAergic neurotransmission, oxidative stress, signal transduction, response to the environment, cell survival and proliferation, and cell shrinkage and apop-
  • 28. tosis, among others [8–13] . Yet no genetic marker has yet been shown to be useful in prospectively identifying any specific psychiatric disorder [14] . Because genetic predis- Despite the burgeoning number of psychiatric treat- ments, we still do not know how to predict which one will work best for which patient. The hope that genetic (single gene effects) and genomic (multiple gene effects) testing might be useful for diagnosis and treatment has been en- couraged by decreased costs of genome sequencing and studies demonstrating an association between mutations in more than 3,000 genes and specific disease phenotypes [1–3] . Are the data as promising in psychiatry as they are in other fields? Cancer Genomics Genetic testing has been most promising in oncology. For example, about 10% of cases of breast cancer have an autosomal dominant pattern of transmission, most com- monly mutations in the tumor suppressor genes BRCA1 and BRCA2 [4] . When BRCA1/2 mutations are found, healthy women are offered a very close follow-up, as well as prophylactic antiestrogen therapy or surgery, yet in one study only 9.5% of high-risk women even underwent genetic counseling, let alone testing [5] . Breast cancer risk alleles have also been found for p53, PTEN (phosphate and tensin homolog deleted from chromosome 10), STK11, CDH1 and PALB2; however, these genetic factors are rare, and there is not much research on screening pro- Received: December 15, 2015 Accepted after revision: December 20, 2015 Published online: April 5, 2016 Steven L. Dubovsky, MD
  • 29. Department of Psychiatry, University at Buffalo 462 Grider Street, Room 1182 Buffalo, NY 14215 (USA) E-Mail dubovsky @ buffalo.edu © 2016 S. Karger AG, Basel 0033–3190/16/0853–0129$39.50/0 www.karger.com/pps http://dx.doi.org/10.1159%2F000443512 Dubovsky Psychother Psychosom 2016;85:129–135 DOI: 10.1159/000443512 130 position in psychiatry is thinly distributed over thou- sands of loci, each contributing a small effect, with con- siderable overlap of brain systems and shared genetic fac- tors [14] , sample size in most association studies has generally been too small to produce meaningful, replica- ble results [1, 15, 16] . In addition, epigenetic and oth- er factors that alter DNA conformation can determine whether susceptibility genes are expressed or suppressed [10] , complicating analyses of the relationship between genotype and phenotype. Even relevant genetic markers can be difficult to interpret because inherited gene factors appear to interact with each other and the environment to contribute to both illness susceptibility and clinical presentation [17, 18] .
  • 30. Descriptive diagnoses in psychiatry have multiple do- mains such as age of onset, constellations of specific symptoms, functioning, comorbidity, and evolution over time that assort differently in different patients in the same category to produce functionally different condi- tions [19] . Genetic profiles associated with any one of these features are not likely to predict more global diag- noses. If the direct path from genotype to phenotype ends at discrete endophenotypes such as arousal, anhedonia, information processing, stress responses, inflammation and mood, rather than global diagnosis, attempts to link the latter to specific genes are likely to prove frustrating [20–23] , just as descriptive diagnoses in psychiatry do not adequately consider important subtypes that exhibit dif- ferent assortments of features such as age of onset, sever- ity, progression or functioning. Genetic Pharmacokinetic Studies Since cytochrome P450 (CYP450) 1A2, 2D6, 2C9, 2C19, and 3A4 account for 60% of psychiatric drug me- tabolism [24] , considerable interest has centered on using the CYP450 genotype to predict response to psychotropic medications [3] . However, genotype does not inevitably predict phenotype because multiple copies of a more or less active gene can result in more or less metabolic activ- ity than would be expected from the allele that is identi- fied. In addition, the metabolizer phenotype associated with a particular genotype can be inhibited or enhanced by a number of medications, substances, and foods [25– 30] . In an open study of 900 patients treated with venla- faxine who were both genotyped and phenotyped for CYP2D6, 4% were genotypically poor metabolizers, while 27% were phenotypically poor metabolizers, suggesting that 23% of patients with other genotypes had converted
  • 31. to a poor metabolizer phenotype as a result of concomi- tant medications [31] . Even if genotype inevitably predicted phenotype, the correlation is stronger between CYP450 phenotype and drug level than clinical response [32] , which is modified by metabolism of most medications by more than one enzyme, lack of linear kinetics and saturable elimination for many drugs, unclear correlations between blood level and response for many medications, and therapeutic win- dows requiring therapeutic monitoring anyway for some of them [24, 26, 33, 34] . Expression of CYP450 enzymes in the brain, which influences drug effect, may be differ- ent from their expression in the blood, or even in the in- testine and liver [24] . Drug Transporter Studies Drug transporters, including P-glycoprotein (P-gp), or- ganic ion transporters, and multidrug and toxin extrusion proteins, modify the effect of CYP450 phenotype on drug levels and drug action because they influence gastrointesti- nal absorption, tissue uptake, and renal elimination as well as transport in and out of the brain [35, 36] . In the iSPOT- D (International Study to Predict Optimized Treatment in Depression), two different MDR1 single nucleotide poly- morphisms (SNPs) of the gene for P-gp (MDR1 or ABCB1) were associated with better responses either to escitalo- pram and sertraline or to venlafaxine, but there was no a priori hypothesis, other relevant factors such as drug me- tabolism, ethnicity, age, specific symptoms, or concomitant illness were not addressed [36] , and DNA was collected af- ter results were known rather than prospectively [37] . Pharmacodynamic Studies
  • 32. Both a deletion (short polymorphism or s-allele) and an insertion (long polymorphism or l-allele) have been found in the gene (SCL6A4) for the promoter region (5-HTTLPR) of the gene for the serotonin transporter (SERT) [32] . The short polymorphism (s) decreases and the long polymorphism (l) increases SLC6A4 transcrip- tion rates, resulting in less or more SERT expression, re- spectively [32] . Research on the association of the s/s ge- notype with a lower response rate to serotonin reuptake inhibitors in some ethnic groups has been contradictory [38–40] , and even without correction for multiple statisti- cal tests, the SCL6A4 genotype explains at most 5% of the variance in antidepressant response [30] . Attempts have http://dx.doi.org/10.1159%2F000443512 Genetics in Psychiatry Psychother Psychosom 2016;85:129– 135 DOI: 10.1159/000443512 131 been made to correlate SNPs of the brain-specific voltage- gated rectifier potassium channel (Kv11.1–3.1) and the cardiac specific version (Kv11.1–1A) with risperidone treatment response and changes in cardiac conduction, respectively, in schizophrenia [41] , but the results have not been robust. Gene Network Studies Because treatment outcome seems to be influenced by multiple genetic polymorphisms, each with a small effect [3, 21] , research has moved toward analysis of networks of genes in the hope of developing more clinically useful
  • 33. information [42–44] . However, such studies have not produced clinically meaningful results [43, 44] . The Ge- nome-Based Therapeutic Drugs for Depression (GEN- DEP; n = 811) study, a substudy of the Sequenced Treat- ment Alternatives to Relive Depression (STAR * D; n = 1,491) study, and the Munich Antidepressant Response Study (n = 339), did not find any combination of genetic markers that influenced treatment response in depres- sion [1, 44] . Genome-wide association studies did not re- veal any SNPs associated with response or remission of nonbipolar, nonpsychotic, major depressive disorder treated openly with serotonin reuptake inhibitors [45] , and the STAR * D study did not reveal any positive ge- nome-wide association or top 25 SNP associations with treatment response [45] . A genome-wide association study from the Clinical Antipsychotic Trials of Interven- tion Effectiveness (CATIE, n = 738) did not find any com- binations of genetic markers that influenced treatment response in schizophrenia [1, 44] . Prospective Treatment Studies Only a small number of reports have involved the pro- spective use of genotyping to make treatment decisions. An open study of 58 depressed inpatients reported that genotyping for ABCB1 was associated with a shorter hos- pital stay because patients with the TT/GG genotype were more likely to have an increase in the dose of an antide- pressant that was a P-gp substrate, although changing to a non-P-gp substrate did not affect outcome [46] . The study was not randomized, and numerous intervening variables, including pharmacokinetics, comorbidity, and history, were not considered. Four studies have been supported by the manufactur- er of a proprietary survey (GeneSight) of CYP2D6, 2C19,
  • 34. 2C9, and 1A2, SLC6A4, and 5HTR2A genotypes that gen- erates a ‘composite report’ classifying antidepressants and antipsychotic drugs used in the treatment of depres- sion into three categories: ‘use as directed’, ‘use with cau- tion’, and ‘use with caution and with more frequent mon- itoring’. An 8-week open study of 44 patients assigned in a nonrandom manner to treatment guided by the com- posite report (guided treatment) or nonguided treatment by the same clinicians, who were involved with the prod- uct, reported that patients in the guided group were less likely to receive medications in the ‘use with caution and with more frequent monitoring’ category, presumably because of reluctance by the guided clinicians to prescribe medications that required more monitoring [47] . Al- though improvement of depression was similar for the first 4 weeks in both groups, a single measure at 8 weeks indicated increased depression scores for the nonguided but not the guided group. No explanation was offered for the final increase in depression scores in the nonguided group, when multiple earlier ratings demonstrated a steady decrease in scores. Improvement in the guided group was not impressive, and it is impossible to know whether comorbid factors, concomitant medications, treatment adherence, patient enthusiasm, substance use, adjunctive psychotherapy, clinician knowledge of treat- ment condition, and open ratings affected the conduct of treatment or the outcome assessment. A second open, nonrandomized study conducted by the same group in 227 mildly-moderately depressed pa- tients, 165 of whom completed 8 weeks of treatment, re- ported that patients in the guided group were twice as likely to respond [48] . Since clinicians reported substan- tial levels of confidence in the genetic reports, it is possi- ble that they worked more vigorously with patients in the
  • 35. guided group, that patients in the guided group were more adherent with a treatment approach they thought would be more effective, or that they reported better re- sults to please the investigators. In a double-blind, randomized, controlled trial of GeneSight, 25 depressed patients were assigned to treat- ment as usual and 26 to guided treatment [49] . Improve- ment was numerically greater in patients in the guided than in the treatment-as-usual group, but none of the group differences were statistically significant. In a fourth report from the same company [50] , 97 patients with a depressive or anxiety disorder treated openly by a single psychiatrist with one of the medications in the genetic sur- vey were followed openly for 1 year. The 9 patients taking at least one medication in the ‘use with caution’ category had significantly more total health care visits and nonpsy- http://dx.doi.org/10.1159%2F000443512 Dubovsky Psychother Psychosom 2016;85:129–135 DOI: 10.1159/000443512 132 chiatric medical visits than the other subjects and had a higher average cost of care. However, these patients also took more medications than the other subjects, and there was a significant correlation between the number of med- ications taken and the two outcome variables. When dif- ferent statistical analyses were performed on the same data (e.g. analysis of variance and t tests), some were sig-
  • 36. nificant and some were not, and no correction was made for multiple statistical tests. Although the authors con- tended that their genetic analysis could save health care costs, this hypothesis was not actually tested. Since no data were available on medical comorbidity and severity of psychiatric illness, the possibility was not considered that the small number of patients in the ‘use with caution’ cat- egory had more health care visits and took more medi- cations because they were sicker psychiatrically or medi- cally. Gene Expression Current genotyping approaches in psychiatry consider the presence or absence of a particular allele or group of alleles, but expression of those genes may be suppressed, modified, or enhanced by a number of factors, including circadian transcription patterns, epistasis (gene interac- tions), regulatory regions, epigenetic factors, and noncod- ing RNA [14] . Histone modification and DNA methyla- tion in response to experience, inflammation, the illness, and the medications used to treat it can induce or suppress multiple genes, and genotype itself can affect methylation of regulatory sites that leads to epigenetic changes in brain development [51] . Micro-RNAs and short interfering RNAs are short, noncoding posttranslational regulators of gene expression that target hundreds of mRNA transcripts to influence gene networks [27, 52, 53] . The expression of genes for CYP450 enzymes is altered by promoter meth- ylation, micro-RNAs associated with inflammation and other illnesses [27] , and some medications [54] , resulting in an altered CYP450 phenotype. Limitations of Pharmacogenetic Testing in Psychiatry
  • 37. No matter how much we may want to translate direct- ly to clinical diagnosis reports of an association between a diagnosis and a genetic marker, the nature of the current level of knowledge does not permit this application. Sam- ple sizes in most existing studies have been too small to produce meaningful, replicable results because of the clinical and genetic heterogeneity of psychiatric disorders [55] , and the combined influence of multiple genes, each with a small effect size [1, 15, 16] . Most studies have uti- lized retrospective or post hoc analyses rather than pro- spective a priori hypotheses [56] , and statistical signifi- cance is often inflated by lack of correction for multiple statistical tests [16] . The majority of studies lack replica- tion in independent samples, especially by different in- vestigators [16] . Even robust findings would not be clini- cally applicable until a prospective study demonstrated their ability to preferentially predict one diagnosis or even clinically relevant feature over another. Using genotype to predict response to medications is even more problematic. Pharmacogenetic studies have been conducted in normal subjects or patients who are not taking other medications and who do not have other illnesses, limiting extrapolation to most clinical settings [33, 34] . Most studies do not control for the effect on the expression of CYP450 and other genes of age [27] , ethnic- ity [30] , smoking [30, 34] , and use of substances such as alcohol, hormones, St. John’s wort, caffeine, cabbage, and grapefruit juice [30] . Genetic studies of treatment out- come have not measured nonadherence [57] , but as the rate of nonadherence increases in any population, statis- tical power to detect a genotype effect decreases substan- tially [1] . For medications that are chiral mixtures of en- antiomers with different actions, the metabolism of each
  • 38. enantiomer may be by different enzymes [58] . Active me- tabolites with their own metabolic pathways may en- hance or interfere with therapeutic or toxic effects pre- dicted by the presumed metabolism of the parent drug [59, 60] . In most instances, more than one genetic factor affects drug levels and disposition [3] , and interactions between these factors can be difficult to predict. A clear demonstration of a genotype/blood level rela- tionship in a single dose or 8-week study may not corre- late with chronic treatment, in which compensatory changes in secondary metabolic pathways and drug trans- porters, gene up- or downregulation, saturation pharma- cokinetics and other factors may modify the impact of oxidative enzyme polymorphisms on final drug level [24, 59] . With chronic treatment, some psychotropic drug metabolites form complexes with P450 enzymes that alter or even reverse the acute effect on metabolism [25] . Long- term changes in P450 enzymes also occur in the brain, with further unpredictable effects, not only on the sub- strate drug, but on neurotransmitters and neurosteroids metabolized by the same enzymes on which the medica- tion may act [25] . Another complicating factor is that the http://dx.doi.org/10.1159%2F000443512 Genetics in Psychiatry Psychother Psychosom 2016;85:129– 135 DOI: 10.1159/000443512 133 illness, as well as medications used to treat it, can alter the relationship between pharmacologic genotype and phe- notype. For example, many proinflammatory cytokines
  • 39. and acute-phase proteins that are associated with mood and anxiety disorders [61] act on transcription or post- translational protein modification to downregulate some CYP450 genes and upregulate others [62] . At the same time, suppression of cytokines by antidepressants can al- ter gene expression in directions that antagonize im- provement of depression [25] . The impact of evolution of the illness and its response to different treatments in modifying therapeutic strategies during the course of treatment of cancer is relatively straightforward to study by virtue of methodologies for examining genotype and phenotype of cellular clones, but it is still difficult to de- velop the correct approach to well-characterized tumors [63] . The absence of such measures in psychiatric diagno- ses makes this prospect considerably more difficult. Where Do We Go from Here? Psychiatrists, whose work frequently involves ambigu- ous clinical problems, and who must often consider con- tradictory elements of patient presentations and avoid premature closure, can have a remarkably low tolerance for ambiguity, conflict, and delayed gratification when it comes to the latest laboratory study. The hope that phar- macogenetic testing will result in unambiguous ‘person- alized psychiatry’ should not lead to quick adoption of technologies that have not yet been demonstrated to reli- ably predict a specific course or a need for a specific med- ication, the choice of which remains largely empirical. Af- ter all, genetic associations are statistical, but medical practice is personal [14] . Yet there is tremendous pressure to translate each new report of such associations to our patients, not only from our own need to appear ‘scien- tific’ and from industry marketing of proprietary tests, but from the marketing of ideas by thought leaders with
  • 40. an intellectual attachment to the latest conceptualization of genetic causality [64] . It is a continuing challenge to examine new genetic findings critically without applying them immediately in the clinic. When adequately powered studies that address gene number and expression and that control for real-life factors that affect outcome such as comorbidity, poly- pharmacy, environmental exposure, age, gender, ethnic- ity, substance use, and treatment adherence emerge [44] , clinicians who have not put new information into action before integrating it with emerging knowledge about di- agnosis, neurobiology, and the evolution of complex dis- orders will be ready to apply them effectively. Disclosure Statement Dr. Dubovsky has received research support from Janssen, Ot- suka, Sumitomo, Neurocrine, Tower Foundation, Wendt Founda- tion, Oshei Foundation and Patrick Lee Foundation. The author has no other conflicts of interest to disclose. References 1 Malhotra AK, Zhang J-P, Lencz T: Pharmaco- genetics in psychiatry: translating research into clinical practice. Mol Psychiatry 2012; 17: 760–769. 2 Cox DBT, Platt RJ, Zhang F: Therapeutic ge- nome editing: prospects and challenges. Nat Med 2015; 21: 121–131. 3 Trembplay J, Hamet P: Role of genomics on
  • 41. the path to personalized medicine. Metabo- lism 2013; 62:S2–S5. 4 Stuckey AR, Onstad MA: Hereditary breast cancer: an update on risk assessment and ge- netic testing in 2015. Am J Obstet Gynecol 2015; 213: 161–165. 5 Bellcross CA, Peipins LA, McCarty FA, Ro- driguez JL, Hawkins NA, Hensley Alford S, Leadbetter S: Characteristics associated with genetic counseling referral and BRCA1/2 test- ing among women in a large integrated health system. Genet Med 2015; 17: 43–50. 6 Ivshina AV, George J, Senko O, Mow B, Putti TC, Smeds J, Lindahl T, Pawitan Y, Hall P, Nordgren H, Wong JEL, Liu ET, Bergh J, Kuznetsove VA, Miller LD: Genetic reclassi- fication of histologic grade delineates new clinical subtypes of breast cancer. Cancer Res 2006; 66: 10292–10301. 7 Anonymous: The future of cancer genomics. Nat Med 2015; 21: 99. 8 Le-Niculescu H, Balaraman Y, Patel SD, Ayalew M, Gupta J, Kuczenski R, Shekhar A, Shekhar A, Schork N, Geyer MA, Niculescu AB: Convergent functional genomics of anxi- ety disorders: translational identification of genes, biomarkers, pathways and mecha- nisms. Transl Psychiatry 2011; 1:e9. 9 Carrard A, Salzmann A, Malafosse A, Karege F: Increased DNA methylation status of the
  • 42. serotonin receptor 5HTR1A gene promoter in schizophrenia and bipolar disorder. J Af- fect Disord 2011; 132: 450–453. 10 Mill J, Tang T, Kaminsky Z, Khare T, Yaz- danpanah S, Bouchard L, Jia P, Assadzadeh A, Flanagan J, Schumacher A, Wang SC, Petro- nis A: Epigenomic profiling reveals DNA- methylation changes associated with major psychosis. Am J Hum Genet 2008; 82: 696– 711. 11 Fullerton JM, Tiwari Y, Agahi G, Heath A, Berk M, Mitchell PB, Schofield PR: Assessing oxidative pathway genes as risk factors for bi- polar disorder. Bipolar Disord 2010; 12: 550– 556. 12 Hoenicka J, Garrido E, Ponce G, Rodriguez- Jimenez R, Martinez I, Rubio G, Jimenez-Ar- riero MA, Palomo T: Sexually dimorphic in- teraction between the DRD1 and COMT genes in schizophrenia. Am J Med Genet 2010; 153B:984–954. http://dx.doi.org/10.1159%2F000443512 Dubovsky Psychother Psychosom 2016;85:129–135 DOI: 10.1159/000443512 134
  • 43. 13 Hong CJ, Liou YJ, Bai Y, Chen TT, Wang Y, Tsai SJ: Dopamine receptor D2 gene is associ- ated with weight gain in schizophrenia pa- tients under long-term atypical antipsychotic treatment. Pharmacogenet Genomics 2010; 20: 359–366. 14 Li JZ: Circadian rhythms and mood: opportu- nities for multi-level analyses in genomics and neuroscience. Bioessays 2013; 36: 305– 315. 15 Genetics of Personality Consortium: Meta- analysis of genome-wide association studies for neuroticism, and the polygenic associa- tion with major depression. JAMA Psychiatry 2015; 72: 642–650. 16 Gillis NK, Innocenti F: Evidence required to demonstrate clinical utility of pharmacoge- netic testing: the debate continues. Clin Phar- macol Ther 2014; 96: 655–657. 17 Fanous AH, Kendler KS: Genetic heterogene- ity, modifier genes, and quantitative pheno- types in psychiatric illness: searching for a framework. Mol Psychiatry 2005; 10: 6–13. 18 Patel SD, Le-Niculescu H, Koller DL, Green SD, Lahiri DK, McMahon FJ, Nurnberger JIJ, Niculescu AB: Coming to grips with complex disorders: genetic risk prediction in bipolar disorder using panels of genes identified through convergent functional genomics. Am J Med Genet 2010; 153B:850–877.
  • 44. 19 Fava GA, Guidi J, Grandi S, Hasler G: The missing link between clinical states and bio- markers in mental disorders. Psychother Psy- chosom 2014; 83: 136–141. 20 Van Borkulo C, Boschioo L, Borsboom D, Pennix BWJH, Waldorp LJ, Schoevers RA: Association of symptom network structure with the course of longitudinal depression. JAMA Psychiatry 2015; 72: 1219–1226. 21 Keers R, Aitchison KJ: Pharmacokinetics of antidepressant response. Expert Rev Neu- rother 2011; 11: 101–125. 22 Domschke K: Clinical and molecular genetics of psychotic depression. Schizophr Bull 2013; 39: 766–775. 23 Fabbri C, Porcelli S, Serretti A: From pharma- cogenetics to pharmacogenomics: the way to- ward the personalization of antidepressant treatment. Can J Psychiatry 2014; 59: 62–75. 24 Preissner SC, Hoffmann MF, Preissner R, Dunkel M, Gewiess A, Preissner S: Polymor- phic cytochrome P450 enzymes (CYPs) and their role in personalized therapy. PLoS One 2013; 8:e82562. 25 Wladyslawa AD: The influence of long-term treatment with psychotropic drugs on cyto- chrome P450: the involvement of different mechanisms. Expert Opin Drug Metab Toxi- col 2005; 1: 203–217.
  • 45. 26 Crews KR, Gaedigk A, Dunnenberger HM, Leeder JS, Klein TE, Caudle KE, Haidar CE, Shen DD, Callaghan JT, Sadhasivam S, Prows CA, Kharasch ED, Skaar TC: Clinical Phar- macogenetics Implementation Consortium guidelines for cytochrome P450 2D6 geno- type and codeine therapy: 2014 update. Clin Pharmacol Ther 2014; 95: 376–382. 27 Zanger UM, Klein K, Thomas M, Rieger JK, Tremmel R, Kandel BA, Klein BA, Klein M, Magdy T: Genetics, epigenetics, and regulation of drug-metabolizing cytochrome p450 en- zymes. Clin Pharmacol Ther 2014; 95: 258–261. 28 Misaka S, Kawabe K, Onoue S, Werba JP, Gi- roli M, Tamaki S, Kan T, Kimura J, Watanabe H, Yamada S: Effects of green tea catechins on cytochrome P450 2B6, 2C8, 2C19, 2D6 and 3A activities in human liver and intestinal mi- crosomes. Drug Metab Pharmacokinet 2013; 28: 244–249. 29 Yamasaki I, Yamada M, Uotsu N, Teramoto S, Takayanagi R, Yamada Y: Inhibitory effects of kale ingestion on metabolism by cyto- chrome P450 enzymes. Biomed Res 2012; 33: 235–242. 30 Jones DS, Perlis RH: Pharmacogenetics, race, and psychiatry: prospects and challenges. Harvard Rev Psychiatry 2006; 14: 92–108. 31 Preskorn SH, Kane CP, Lobello K, Nichols AI, Fayyad R, Buckley G, Focht K, Guico-Pabia CJ: Cytochrome P450 2D6 phenoconversion
  • 46. is common in patients being treated for de- pression: implications for personalized medi- cine. J Clin Psychiatry 2013; 74: 614–621. 32 Alter CN, Hornberger J, Shewade A, Cruz V, Garrison J, Mrazek D: Clinical validity of cy- tochrome P450 metabolism and serotonin gene variants in psychiatric pharmacothera- py. Int Rev Psychiatry 2013; 25: 509–533. 33 De Leon J, Arranz GR: Pharmacogenetic test- ing in psychiatry: a review of features and clin- ical realities. Clin Lab Med 2008; 28: 599–617. 34 Kirchheiner J, Brosen K, Dahl ML, Gram LF, Kasper S, Roots I, Sjoqvist F, Spina E, Brock- moller J: CYP2D6 and CYP2C19 genotype- based dose recommendations for antidepres- sants: a first step towards subpopulation-spe- cific dosages. Acta Psychiatr Scand 2001; 104: 173–192. 35 Rosenhagen MC, Uhr M: The clinical impact of ABCB1 polymorphisms in the treatment of psychiatric diseases. Curr Pharm Des 2011; 17: 2843–2851. 36 Goswami S, Yee SW, Stocker S, Mosley JD, Kubo M, Castro R, Mefford JA, Wen C, Liang X, Witte J, Brett C, Maeda S, Simpson MD, Hedderson MM, Davis RL, Roden DM, Gia- comini KM, Savic RM: Genetic variants in transcription factors are associated with the pharmacokinetics and pharmacodynamics of metformin. Clin Pharmacol Ther 2014; 96: 370–370.
  • 47. 37 Schatzberg AF, DeBattista C, Lazzeroni LC, Etkin A, Murphy GM, Williams LM: ABCB1 genetic effects on antidepressant outcomes: a report from the iSPOT-D trial. Am J Psychia- try 2015; 172: 751–759. 38 Porcelli S, Fabbri C, Serretti A: Meta-analysis of serotonin transporter gene promoter poly- morphism (5-HTTLPR) association with an- tidepressant efficacy. Eur Neuropsychophar- macol 2012; 22: 239–258. 39 Taylor MJ, Sen S, Bhagwagar Z: Antidepres- sant response and the serotonin transporter- linked polymorphic region. Biol Psychiatry 2010; 68: 536–543. 40 Kang HJ, Kim JM, Stewart R, Kim SY, Bae KY, Kim SW, Shin MG, Yoon JS: Association of SLC6A4 methylation with early adversity, characteristics and outcomes in depression. Prog Neuropsychopharmacol Biol Psychiatry 2013; 44: 23–28. 41 Heide J, Zhang F, Bigos KL, Mann SA, Carr VJ, Weickert CS, Green MJ, Weinberger DR, Vandenberg JI: Differential response to risperidone in schizophrenia patients by KCNH2 genotype and drug metabolizer sta- tus. Am J Psychiatry 2016; 173: 53–59. 42 Costa e Silva JA: Personalized medicine in psychiatry: new technologies and approaches. Metabolism 2013; 62:S40–S44.
  • 48. 43 Singh AB, Bousman CA, Ng C, Berk M: Anti- depressant pharmacogenetics. Curr Opin Psychiatry 2014; 27: 43–51. 44 Hamilton SP: The promise of psychiatric pharmacogenomics. Biol Psychiatry 2015; 77: 29–35. 45 Ji Y, Biernacka JM, Hebbring S, Jenkins GD, Batzler A, Snyder KA, Drews MS, Desta Z, Flockart D, Mushiroda T, Kubo M, Naka- mura Y, Kamatani N, Schaid D, Weinshil- boum RM, Mrazek D: Pharmacogenomics of selective serotonin reuptake inhibitor treatment for major depressive disorder: ge- nome-wide associations and functional ge- nomics. Pharmacogenomics J 2013; 13: 456– 463. 46 Breitenstein B, Scheuer S, Pfister H, Uhr M, Lucae S, Holsboer F, Ising M, Bruckl T: The clinical application of ABCB1 genotyping in antidepressant treatment: a pilot study. CNS Spectr 2014; 19: 165–175. 47 Hall-Flavin DK, Winner JG, Allen J, Jordan JJ, Nesheim RS, Snyder KA, Drews MS, Eister- hold LL, Biernacka JM, Mrazek D: Using a pharmacogenomic algorithm to guide the treatment of depression. Transl Psychiatry 2012; 2:e172. 48 Hall-Flavin DK, Winner JG, Allen J, Carhart JM, Proctor B, Snyder KA, Drews MS, Eister- hold LL, Geske J, Mrazek D: Utility of inte- grated pharmacogenomic testing to support
  • 49. the treatment of major depressive disorder in a psychiatric outpatient setting. Pharmaco- genet Genomics 2013; 23: 535–548. 49 Winner JG, Carhart JM, Alter CN, Allen J, Dechairo BM: A prospective, randomized, double-blind study assessing the clinical im- pact of integrated pharmacogenomic testing for major depressive disorder. Discov Med 2013; 16: 219–227. 50 Winner JG, Allen J, Altar CA, Spahic-Miha- jlovic A: Psychiatric pharmacogenomics pre- dicts health resource utilization of outpatients with anxiety and depression. Transl Psychia- try 2013; 3:e242. http://dx.doi.org/10.1159%2F000443512 Genetics in Psychiatry Psychother Psychosom 2016;85:129– 135 DOI: 10.1159/000443512 135 51 Hannon E, Spiers H, Viana J, Pidsley R, Bur- rage J, Murphy TM, Troakes C, Turecki G, O’Donovan MC, Schalkwyk LC, Bray NJ, Mill J: Methylation QTLS in the developing brain and their enrichment in schizophrenia risk loci. Nat Neurosci 2016; 19: 48–54. 52 Geaghan M, Cairns MJ: MicroRNA and post- transcriptional dysregulation in psychiatry. Biol Psychiatry 2015; 78: 231–239.
  • 50. 53 Xu Q, Wu X, Xiong Y, Xing Q, He L, Qin S: Pharmacogenomics can improve antipsy- chotic treatment in schizophrenia. Front Med 2013; 7: 180–190. 54 Gardiner E, Carroll A, Tooney PA, Cairns MJ: Antipsychotic drug-associated gene-miRNA interaction in T-lymphocytes. Int J Neuro- psychopharmacol 2014; 17: 929–943. 55 Le-Niculescu H, Kurian SM, Yehyawi N, Dike C, Patel SD, Edenberg HJ, Tsuang MT, Salo- mon DR, Nurnberger JIJ, Niculescu AB: Iden- tifying blood biomarkers for mood disorders using convergent functional genomics. Mol Psychiatry 2009; 14: 156–174. 56 Janssens ACJW, Deverka PA: Useless until proven effective: the clinical utility of pre- emptive pharmacogenetic testing. Clin Phar- macol Ther 2014; 96: 652–654. 57 Melfi CA, Chawla AJ, Croghan TW, Hanna MP, Kennedy S, Sredl K: The effects of adher- ence to antidepressant treatment guidelines on relapse and recurrence of depression. Arch Gen Psychiatry 1998; 55: 1128–1132. 58 Crettol S, de Leon J, Hiemke C, Eap CB: Phar- macogenomics in psychiatry: from therapeu- tic drug monitoring to genomic medicine. Clin Pharmacol Ther 2014; 95: 254–257. 59 Kirchheiner J, Nickchen K, Bauer M, Wong M-L, Roots I, Brockmoller J: Pharmacogenet-
  • 51. ics of antidepressants and antipsychotics: the contribution of allelic variations to the phe- notype of drug response. Mol Psychiatry 2004; 9: 442–473. 60 Seeringer A, Kirchheiner J: Pharmacogenet- ics-guided dose modifications of antidepres- sants. Clin Lab Med 2008; 28: 619–626. 61 Young JJ, Bruno D, Pomara N: A review of the relationship between proinflammatory cyto- kines and major depressive disorder. J Affect Disord 2014; 169: 15–20. 62 Harvey RD, Morgan ET: Cancer, inflamma- tion, and therapy: effects on cytochrome p450-mediated drug metabolism and impli- cations for novel immunotherapeutic agents. Clin Pharmacol Ther 2014; 96: 449–457. 63 Michor F, Beal K: Improving cancer treat- ment via mathematical modeling: surmount- ing the challenges is worth the effort. Cell 2015; 163: 1059–1063. 64 Dubovsky SL, Dubovsky AN: Psychotropic Drug Prescriber’s Survival Guide: Ethical Pre- scribing in the Age of Big Pharma. New York, Norton, 2007. http://dx.doi.org/10.1159%2F000443512 CitRef_1: CitRef_2: CitRef_3: CitRef_4: CitRef_5: CitRef_6: CitRef_7: CitRef_8: CitRef_9: CitRef_10: CitRef_11: CitRef_12: CitRef_13: CitRef_14: CitRef_15: CitRef_16:
  • 52. CitRef_17: CitRef_18: CitRef_19: CitRef_20: CitRef_21: CitRef_22: CitRef_23: CitRef_24: CitRef_25: CitRef_26: CitRef_27: CitRef_28: CitRef_29: CitRef_30: CitRef_31: CitRef_32: CitRef_33: CitRef_34: CitRef_35: CitRef_36: CitRef_37: CitRef_38: CitRef_39: CitRef_40: CitRef_41: CitRef_42: CitRef_43: CitRef_44: CitRef_45: CitRef_46: CitRef_47: CitRef_48: CitRef_49: CitRef_50: CitRef_51: CitRef_52: CitRef_53: CitRef_54: CitRef_55: CitRef_56: CitRef_57: CitRef_58: CitRef_59: CitRef_60: CitRef_61: CitRef_62: CitRef_63: