Você está na página 1de 8

Journal of Psychiatric Research 96 (2018) 100e107

Contents lists available at ScienceDirect

Journal of Psychiatric Research


journal homepage: www.elsevier.com/locate/psychires

Improved efficacy with targeted pharmacogenetic-guided treatment


of patients with depression and anxiety: A randomized clinical trial
demonstrating clinical utility
Paul Bradley a, h, Michael Shiekh b, Vishaal Mehra c, Keith Vrbicky d, e, f, Stacey Layle c,
Marilyn C. Olson g, Alejandra Maciel g, *, Ali Cullors g, Jorge A. Garces g,
Andrew A. Lukowiak g
a
BCG Medical Group, Internal Medicine, Mercer University School of Medicine, Savannah, GA, United States
b
Relaro Medical Trials, Dallas, TX, United States
c
Artemis Institute for Clinical Research, San Diego, CA, United States
d
Midwest Health Partners, P.C., Norfolk, NE, United States
e
Creighton University, Department of OB/GYN, United States
f
University of Nebraska Medical Center, Department of OB/GYN, United States
g
AltheaDx, San Diego, CA, United States
h
Meridian Clinical Research, Savannah, GA, United States

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

Article history: The objective of this study was to evaluate the effect of pharmacogenetics-guided treatment on patients
Received 24 June 2017 diagnosed with depression and/or anxiety, in a diverse set of clinical settings, as compared to the
Received in revised form standard of care. The trial design followed a prospective, randomized, subject- and rater-blinded
22 August 2017
approach enrolling 685 patients from clinical providers specializing in Psychiatry, Internal Medicine,
Accepted 22 September 2017
Obstetrics & Gynecology, and Family Medicine. The NeuroIDgenetix® test uses a genetic variant panel of
ten genes, along with concomitant medications, to make medication management recommendations
Keywords:
based on gene-drug and drug-drug interactions for over 40 medications used in the treatment of
Pharmacogenetics
Depression
depression and anxiety. Pharmacogenetic testing was performed at the initial screening visit and
Anxiety baseline patient assessments were determined using the 17-item Hamilton Rating Scale for Depression
Efficacy (HAM-D17) and the Hamilton Rating Scale for Anxiety (HAM-A). Following enrollment and randomi-
Treatment response zation, pharmacogenetic results for subjects assigned to the experimental group were provided to
Precision medicine physicians to guide treatment selection, while control subjects were treated according to the usual
standard of care. HAM-D17 and HAM-A assessments were collected at 4 weeks, 8 weeks, and 12 weeks
after baseline to assess the efficacy of therapeutic selection. In patients diagnosed with depression,
response rates (p ¼ 0.001; OR: 4.72 [1.93e11.52]) and remission rates (p ¼ 0.02; OR: 3.54 [1.27e9.88])
were significantly higher in the pharmacogenetics-guided group as compared to the control group at 12
weeks. In addition, patients in the experimental group diagnosed with anxiety showed a meaningful
improvement in HAM-A scores at both 8 and 12 weeks (p ¼ 0.02 and 0.02, respectively), along with
higher response rates (p ¼ 0.04; OR: 1.76 [1.03e2.99]). From these results, we conclude that
pharmacogenetic-guided medication selection significantly improves outcomes of patients diagnosed
with depression or anxiety, in a variety of healthcare settings.
© 2017 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

1. Introduction

Approximately 16 million Americans suffer from moderate or


severe depression, a medical condition that affects mood, cognitive
* Corresponding author. AltheaDx, 10578 Science Center Drive, San Diego, CA and physical symptoms (Pratt and Brody, 2014). The costs of in-
92121, United States.
dividuals with Major Depressive Disorder is greater than $210
E-mail address: AMaciel@altheadx.com (A. Maciel).

https://doi.org/10.1016/j.jpsychires.2017.09.024
0022-3956/© 2017 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
P. Bradley et al. / Journal of Psychiatric Research 96 (2018) 100e107 101

billion annually including direct costs such as medical and phar- 95% CI 1.71e3.73, N ¼ 148) (Singh, 2015). While informative, the
maceutical services in addition to workplace costs (Greenberg et al., narrow patient population and the low sample number of these
2015). One in eleven US adults reports the use of an antidepressant studies limits the applicability of the results to the general popu-
medication in the past 30 days, making antidepressants the third lation. For example, both Winner and Singh exclude patients with
most commonly prescribed drug type (US Dept. Health and Human comorbid psychiatric disorders and enroll patients only seen at
Services, Centers for Disease Control and Prevention, 2013). Despite psychiatric clinics. Given the high number of non-psychiatric cli-
their broad use in treating depression, initial success in medication nicians prescribing anti-depressants and the high rate of comor-
selection can be low. Findings from the Sequenced Treatment Al- bidity for depression and anxiety, a clinical study examining the
ternatives to Relieve Depression (STAR*D) study demonstrated that clinical utility of pharmacogenetics in depression patients should
only one-third of the patients achieved remission during the first aim to reflect the real-world clinical setting.
treatment level (Trivedi et al., 2006). Furthermore, patients with The present study utilizes the NeuroIDgenetix® pharmacoge-
difficult-to-treat depression may achieve remission after trying netic test to detect and interpret the impact of an individual pa-
several treatment strategies, but the odds diminish with every tient's genetic variants associated with pharmacokinetic and
additional treatment strategy demonstrating the limitations of pharmacodynamic response, as well as metabolic interactions
trial-and-error medication management (Trivedi et al., 2006; related to concurrent medications, over the counter drugs, herbal
Huynh and McIntyre, 2008). supplements, and diet. The NeuroIDgenetix® Test uses a genetic
While often overshadowed by depression, anxiety disorders are variant panel of ten genes, along with concomitant medications, to
the most prevalent mental disease in the United States with an make medication management recommendations based on gene-
estimated 40 million affected adults age 18 or older and account for drug and drug-drug interactions for over 40 medications used in
more than $42 billion in annual healthcare costs due to frequent the treatment of depression and anxiety (see Supplemental Table 1
medical evaluations and treatment of physical manifestations of and Supplemental Table 2). This study was designed to determine
the disorder (Bystritsky, 2006; Anxiety and Depression Association whether incorporation of the pharmacogenetic test report can
of America, 2016). Approximately one-third of those suffering from improve outcomes in the treatments of anxiety and depression and
an anxiety disorder receive treatment with only about 60% of those address shortcomings of current prescribing practices. Specifically,
patients responding to treatments (Bystritsky, 2006). Further this study compares the therapeutic outcomes of pharmacogenetic-
complicating the treatment of anxiety is that it is frequently part of guided treatment in patients diagnosed with depression or anxiety
a dual diagnosis with depression (comorbidity estimated at 50%), with standard of care medication management.
decreasing the odds of remission and increasing the response time
to pharmacotherapy (Anxiety and Depression Association of 2. Methods
America, 2016; Wittchen et al., 2002; Kessler et al., 2010; Fava
et al., 2008). 2.1. Study design
Therapeutic selection today relies on an empirical approach
using physician experience in combination with clinical indicators This multicenter study (registered at www.clinicaltrials.gov
to determine treatment options, resulting in a “trial-and-error” under identifier number NCT02878928) was designed to deter-
approach to medication management (Mrazek, 2010; Rush et al., mine if pharmacogenetics can help guide therapeutic decisions for
2006). In contrast, pharmacogenetic testing can help reduce the patients with depression and/or anxiety disorders and improve
uncertainty inherent in psychiatric pharmacotherapy by detecting patient outcomes by maximizing drug efficacy. The trial was con-
genetic factors that predict clinical response and side effects, such ducted across 20 independent clinical sites within the US special-
as genetic variations that impact drug-metabolizing enzymes, drug izing in Psychiatry, Internal Medicine, Obstetrics and Gynecology,
transporters or drug targets (Evans and Relling, 1999; Evans and and Family Medicine. Study participants were randomized by dis-
Johnson, 2001; Altar et al., 2013; Porcelli et al., 2011). These asso- ease and severity and allocated in a 1:1 ratio to the experimental
ciations are well supported in the scientific literature and are group (guided by the NeuroIDgenetix® test) or control group
becoming prevalent in professional guidelines and drug labeling (standard of care). For patients within the experimental group, the
(US Food and Drug Administration, 2016; Caudle et al., 2014; Hicks NeuroIDgenetix® test report was released to participating clinicians
et al., 2015). In fact, the American Psychiatric Association guidelines to consider the recommendations from the report when making
for the treatment of major depressive disorder acknowledge indi- medication decisions. For the control group, patients were sub-
vidual pharmacokinetic and pharmacodynamic differences among jected to sample collection procedures identical to the experi-
individuals that may necessitate doses higher than those approved mental group, however NeuroIDgenetix® results were withheld
by the FDA to achieve therapeutic benefit (Gelenberg et al., 2010). and clinicians were asked to manage control subjects according to
Additionally, FDA guidance for new drug approvals further sup- the standard of care. Test results were provided before the first
ports the importance of pharmacogenetic testing to reduce adverse study visit at 2 weeks (±1 week), to guide medication changes in
drug events (ADEs) and personalize dosing (US Food and Drug the experimental group. Medication changes for both groups
Administration, 2013). occurred by the 2 week visit. Patients were monitored and assessed
Previous studies have explored the clinical impact of pharma- for depression and anxiety symptoms using HAM-A and HAM-D17
cogenetic testing in the management of patients diagnosed with interviews by trained raters at study initiation (baseline) and at the
depression. In a review of 465 published records conducted by 4, 8, and 12 week follow-up visits. All patients and raters were
Helfand et al., in 2016, two randomized clinical trials were identi- blinded to the patient's assigned study group until the end of the
fied that compared the effectiveness of pharmacogenetic-testing to trial. Medication changes in both groups (new medications,
standard of care (Helfand et al., 2016). Winner et al., found discontinuation, and dose adjustments) and prescription use were
improved remission rates in patients with previous failed treat- collected at all study visits, as were the occurrence of ADEs, hos-
ments but the results did not reach statistical significance (Gene- pitalizations, medical visits, disability, and missed days of work.
sight: 20% vs 8%, RR 2.40, N ¼ 51) (Winner et al., 2013). Singh
reported improved remission rates in a pharmacogenetic-guided 2.2. Study Participants
group compared to standard of care among Australian patients
with baseline HAM-D scores of 25 (CNSDose: 72% vs. 28%; RR 2.52, A total of 764 patients were assessed for eligibility, 79 subjects
102 P. Bradley et al. / Journal of Psychiatric Research 96 (2018) 100e107

were excluded resulting in the enrollment and randomization of HTR2A [NM_000621.4:c.614e2211T > C], MTHFR
685 patients for the study (Fig. 1). Study participants included pa- [NM_005957.4:c.665C > T], MTHFR [NM_005057.3:c.1286A > C] (see
tients between the ages of 19 and 87 with a diagnosis of depression Supplemental Table 1). The variants tested represent an ethnically-
and/or anxiety using the DSM-V criteria or standard of care site diverse set of genetic polymorphisms, appropriate for the US pop-
procedures and verified by the MINI Psychiatric Interview (Sheehan ulation (see Supplemental Table 1). Molecular methods used for
et al., 1998). Participants were either ‘New to Treatment’ (newly detection included end-point PCR, real-time PCR for CYP2D6 copy
diagnosed or taking medications for less than 6 weeks) or ‘Inade- number variation determination, and capillary electrophoresis for
quately Controlled’ with medications as defined by lack of efficacy the genotyping of SLC6A4 5-HTTLPR (Pratt et al., 2010; Lesch et al.,
or treatment discontinuation due to adverse events or intolera- 1996). In the case of CYP genes, genotypes are translated into dip-
bility. Exclusion criteria consisted of a concurrent diagnosis of bi- lotypes and then diplotypes are assigned a metabolic phenotype for
polar disorder, schizophrenia, personality disorder, traumatic clinical management. As an example, a subject homozygous for the
physical injury (i.e., traumatic brain injury), significant risk for CYP2D6 1846G > A variant, would be assigned a CYP2D6*4/*4 dip-
suicide and hospitalization, patients with a history of chronic renal lotype and a corresponding “Poor Metabolizer” phenotype for the
dysfunction or Chronic Kidney Disease (Stage 4 or 5), malabsorp- purpose of medication selection. Complementing this pharmacoge-
tion (short gut syndrome), pregnancy, or abnormal hepatic function netic analysis, the IDgenetix® algorithm also screens for potential
(INR >1.2 not attributable to anticoagulant medications, AST/ metabolic interactions between concomitant medications as well as
aspartate aminotransferase or ALT/alanine aminotransferase >1.5 a variety of lifestyle factors; including the use of alcohol, tobacco, and
normal, or suspected cirrhosis). This study was carried out in herbal supplements (see Supplemental Fig. 1 for an example of the
accordance with the Declaration of Helsinki, approved by an NeuroIDgenetix® Report).
institutional review board (Aspire IRB) and all participants signed NeuroIDgenetix® reports were sent to the treating physicians,
an approved consent form prior to initiating study activities. All who were blind to the patients' Hamilton Scores, for patients in the
subjects were monitored over a 12 week period. experimental group within approximately 3 days after sample
receipt by the laboratory. The NeuroIDgenetix® report classifies
therapeutically related medications as one of two options; “Use as
2.3. Pharmacogenetic testing Directed” (UAD) or “Use with Caution and/or Increased Monitoring”
(UWC). The medications classified as “Use as Directed” may be
Buccal swabs were collected from all patients prior to randomi- administered in accordance to the manufacturer's standard pre-
zation and shipped to AltheaDx (San Diego, CA) for processing. scribing information since among the genes analyzed and current
Samples were processed in accordance with the Centers for Medi- drug regimen, no genetic variants or metabolic interactions were
care and Medicaid Services (CMS) regulations in a College of Amer- identified that would suggest a need for increased caution or dose
ican Pathologists (CAP)-accredited, Clinical Laboratory Improvement adjustment. Medications classified as “Use with Caution and/or
Amendments (CLIA)- certified laboratory (AltheaDx). In summary, Increased Monitoring” have been identified by the IDgenetix® al-
genomic DNA was extracted from buccal samples using the Qiagen gorithm as having one or more reasons for avoidance, including
DSP DNA Midi Kit (Qiagen, Valencia, CA, USA). Pharmacokinetic and possible drug-gene and drug-drug interactions. These warnings are
Pharmacodynamic variants associated with 10 genes were tested, accompanied by brief descriptions of the reasons for caution (e.g.,
including CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4, CYP3A5, lack of efficacy or toxicity) along with recommendations for
SLC6A4 [NM_001045.5:c.-1760C > T], SLC6A4 [5-HTTLPR], COMT appropriate clinical action.
[NM_000754.3:c.472G > A], HTR2A [NM_000621.4:c.-998G > A],

Fig. 1. Study participants.


P. Bradley et al. / Journal of Psychiatric Research 96 (2018) 100e107 103

2.4. Patient assessments 3. Results

Patients diagnosed with depression were graded using the 17 3.1. Participant characteristics
item Hamilton Depression Rating Scale (HAM-D17) to determine
depression severity at baseline and at the 4, 8 and 12 week follow- A total of 764 patients diagnosed with depression and/or anxi-
up visits (Hamilton, 1960; Cusin et al., 2010). The HAM-D17 inter- ety were eligible to participate in this study of which, 79 were
view was administered by trained clinicians blinded to treatment excluded due to not meeting inclusion/exclusion criteria or
decisions and patient status throughout the study. Total scores declining participation (Fig. 1). The remaining 685 patients were
were categorized as of 0e7 normal, 8e17 mild depression, 18e24 randomized and allocated to either the experimental group
moderate depression, and >24 severe depression. Patients diag- (n ¼ 352) or the control group (n ¼ 333) and followed for period of
nosed with anxiety were assessed for symptoms and severity of up to 12 weeks. At baseline and each follow-up visit (4, 8 and 12
anxiety at baseline and at the 4, 8 and 12 week follow-up visits weeks) patients were assessed for depression and/or anxiety
using the Hamilton Rating for Anxiety (HAM-A) (Hamilton, 1959; symptoms. Patients were allowed to miss up to one visit and
Goodman, 2004). Similar to the HAM-D17 assessments, the HAM- remain in the study. A total of 579 patients (297 experimental, 282
A interviews were conducted by trained and blinded clinicians control) completed the study.
throughout the study. Total scores of 8e17 indicated mild anxiety, Study participants had a mean age of 48 years, with approxi-
18e24 moderate anxiety and >24 severe anxiety. mately 73% female and 27% male, approximating a gender bias
distribution consistent with other reports (Table 1). Patients were
categorized into three different diagnosis categories: depression
2.5. Adverse drug events
(n ¼ 246), anxiety (n ¼ 235) and both depression and anxiety
(n ¼ 204). HAM-D17 assessments were conducted for patients
The Adverse Drug Event form used in this clinical study was
diagnosed with depression and depression and anxiety. HAM-A
developed and reviewed by external clinical psychiatric consultants
assessments were conducted for patients with anxiety and pa-
and used uniformly by all participating sites. The form is divided
tients with both depression and anxiety. Baseline HAM-D17 and
into two sections. The first section is completed during a face to face
HAM-A scores were similar between the experimental and control
interview by the study coordinator. The type of ADE (e.g. nausea,
groups as shown in Table 1. The distribution of metabolizer phe-
dry mouth, weight gain), suspected drug and start date as well as
notypes, based on Clinical Pharmacogenetics Implementation
the event onset and end date (if applicable) are collected. The
Consortium (CPIC) guidelines (Whirl-Carrillo et al., 2012), and other
second section is completed by a blinded clinician and involves
genotypes were statistically indistinguishable between the exper-
grading the seriousness of the ADE on a scale of mild, moderate,
imental and control groups (Supplemental Table 3).
severe, life-threatening or death, and documenting the manage-
ment and outcome of the ADE. Lastly, the clinician evaluates the
likelihood that the suspected drug was the actual cause of the
3.2. Depression outcomes
adverse event. The assessment of causality is rated as one of the
following: unrelated, unlikely, possibly, probably or definitely
To determine whether NeuroIDgenetix®-guided treatment can
related to the suspected drug. All ADE forms were completed by
improve outcomes for patients diagnosed with moderate or severe
trained and blinded clinicians and completed at study enrollment
depression at baseline, we compared HAM-D17 scores between the
and at each follow-up visit.
experimental and control groups at 4, 8 and 12 weeks of follow-up.
Remission rates at the 8 week follow-up visit for experimental
2.6. Statistical analysis patients with severe depression were 25%, whereas the control
group remission rates were 9% (Fig. 2a). At the 12 week visit,
This study was designed to determine the clinical utility of remission rates for experimental patients with severe depression
pharmacogenetics-guided treatment (experimental group) as were 35%, 95% CI [21%, 52%], whereas the control group remission
compared to standard of care treatment (control group) by rates were 13%, 95% CI [5%, 25%], p ¼ 0.02, OR: 3.54 (1.27e9.88),
measuring the changes in the severity of depression and anxiety NNT ¼ 4.6 (Fig. 2a).
symptoms. The efficacy endpoints included HAM-D17 changes for Response rates were significantly higher for patients in the
patients with depression and HAM-A changes for patients with experimental group as well. At the 8 week follow-up visit, the
anxiety measured at baseline and the 4, 8, and 12 week follow-up response rate was higher in the experimental group compared to
visits. Per the study protocol, only those patients with moderate the control group (55% versus 28% respectively) for patients diag-
or severe Hamilton scores (18) at baseline were included in the nosed with severe depression (Fig. 2b). At the 12 week follow-up
efficacy analysis. HAM-D17 and HAM-A scores from patients with visit, response rates trended even higher in the experimental
both depression and anxiety were included in both the HAM-D17 group (73%, 95% CI [56%, 85%]) as compared to the control group
and HAM-A analyses. The impact of pharmacogenetics guided (36%, 95% CI [23%, 50%], p ¼ 0.001, OR: 4.72 (1.93e11.52),
treatment on response rates (50% reduction in HAM scores) and NNT ¼ 2.7). When both moderate and severe patients were
remission rates (HAM scores 7) were evaluated using Fisher's included in the analysis (HAM-D17  20), 8 week response rates
exact test and logistic regression analyses were used to compare were 49% for the experimental group and 41% for the control group.
response and remission rates between the experimental group and The 12 week follow-up visit response rates were significantly
control group. Response and remission rates were defined and higher for patients in the experimental group compared to the
calculated as a subject achieving response or remission at any time control group, 64%, 95% CI [55%, 72%], vs. 46%, 95% CI [37%, 56%],
point after baseline. Changes in the HAM-D17 and HAM-A scores p ¼ 0.01, OR:2.03 (1.23e3.33), NNT ¼ 5.8 (Fig. 2c).
were compared between the experimental and control groups by t- Our study did not find significant improvement in patients with
test and also in a regression analysis accounting for baseline scores mild depression. This finding aligns with research suggesting that
and comorbidity of depression and anxiety. In addition, a mixed patients with mild depression may not benefit from anti-
model repeated measures (MMRM) analysis was performed using depressant medication (Barbui et al., 2011).
all available data from baseline to the 12 week visit.
104 P. Bradley et al. / Journal of Psychiatric Research 96 (2018) 100e107

Table 1
Demographic and clinical characteristics of study participants.

Experimental Group (N ¼ 352) Control Group (N ¼ 333)

Age
Years 47.8 (±14.5) 47.3 (±15.2)
Gender
Female 257 [73%] 241 [72%]
Male 95 [27%] 92 [28%]
Ethnicity
African-American 63 [18%] 59 [18%]
Asian 5 [1%] 4 [1%]
Caucasian 221 [63%] 209 [63%]
Hispanic 57 [16%] 58 [17%]
Other 6 [2%] 3 [1%]
Total Population
Depression (n ¼ 450) 237 213
Baseline HAM-D17 Mean (S.D) 20(±5.8) 20(±5.6)
Baseline HAM-D17 Median 20 20
Anxiety (n ¼ 439) 219 220
Baseline HAM-A Mean (S.D) 25(±7.4) 25(±7.1)
Baseline HAM-A Median 25 24
Moderate and Severe Population(s)
Depression HAM-D17  25 (n ¼ 93) 40 53
Depression HAM-D17  20 (n ¼ 261) 140 121
Anxiety HAM-A 18 (n ¼ 396) 200 196
Anxiety Only HAM-A  18 (n ¼ 224) 109 115

Out of 352 subjects in the experimental group, 133 were classified as depression only, 115 were classified as anxiety only, 104 were classified as depression
and anxiety, leading to a total of 237 (133 þ 104) patients in the experimental arm of depression and 219 (115 þ 104) in the experimental arm of anxiety. Out
of 333 subjects in the control group, 113 were classified as depression only, 120 were classified as anxiety only, 100 were classified as depression and anxiety,
leading to a total of 213 (113 þ 100) patients in the control arm of depression and 220 (120 þ 100) in the control arm of anxiety.

3.3. Anxiety outcomes similar for subjects between groups (p ¼ 0.70). Medication changes
in the experimental group were aligned with the report recom-
The impact of NeuroIDgenetix® testing on guiding therapy for mendations 70% of the time, as opposed to random selection in the
patients diagnosed with anxiety was also evaluated in this study. control group, which aligned with report recommendations
Patients included in this analysis were classified as having moder- (withheld from the treating physician) only 29% of the time. Ulti-
ate to severe anxiety based on HAM-A scores of 18. Anxiety or mately, 83% of the subjects in the experimental group had at least
both depression/anxiety subjects with medication therapy guided one medication concordant with the report. Overall treatment
by the NeuroIDgenetix® test showed a reduction in anxiety changes from baseline were comparable between the experimental
symptoms compared to the control group (Table 2). and control groups, supporting our conclusion that report recom-
The percent reduction in HAM-A scores was even more signif- mendations were highly informative as the experimental group
icant when patients with anxiety only were analyzed. The percent experienced significantly better efficacy over the control.
reduction in HAM-A scores at the 4, 8 and 12 week follow-up visits
within this cohort were 39%, 47% and 54% for the experimental 3.5. Adverse drug events
group versus 26%, 35% and 42% for the control group (p ¼ 0.004,
0.02 and 0.02) (Table 2). Furthermore, at the 12 week follow-up We did not find any statistical difference in ADEs between the
visit, the response rate was significantly higher in the experi- control and the experimental group (p-value ¼ 0.21). Ninety-four
mental group (63%, 95% CI [54%, 72%]) compared to the control percent of total ADEs reported by both groups were not severe,
group (50%, 95% CI [40%, 59%], p ¼ 0.04, OR: 1.76 (1.03e2.99), and included ADEs such as drowsiness, nausea and dry mouth. Of
NNT ¼ 7.3) for patients diagnosed with anxiety only (Fig. 3). the total number of ADEs, 6% were severe and were equally
distributed across the control and experimental groups. The fre-
3.4. Medication changes quency of severe ADEs was too low for statistical comparison.

Physicians implemented or modified therapy two weeks after 4. Discussion


patient enrollment (±1 week). All depression and anxiety medi-
cation changes were recorded along with a list of prescription Diagnoses of anxiety and depression have been increasing
medication use. Medication changes were categorized as one of the rapidly in the US over the last several decades, with the lifetime
following: a new medication added, dose adjustment, or current prevalence of a major depressive disorder near 17% and anxiety
medication discontinued. The number of medication changes disorders affecting 29% of the population (Kessler et al., 2005). The
occurring in the experimental group was significantly higher than U.S. Preventive Services Task Force recommends screening for
the control group during the 2e12 week follow-up visits depression in the general adult population and includes effective
(p < 0.0001, Table 3). Most medication changes occurred at the 2 treatment as part of the implementation plan (Siu and US
week follow up visit; 81% of the subjects in the experimental group Preventative Services Task Force, 2016). Consequently, the re-
versus 64% of subjects in the control group along with additional sponsibility of treating neuropsychiatric illness is no longer falling
changes throughout the study. Within the experimental group, on psychiatrists alone but a wide spectrum of health care providers.
there were more subjects with new medications added and med- This paradigm shift is already well under way as psychiatrists are
ications discontinued, as compared to the control group (p < 0.0001 only responsible for ~20% of the antidepressants prescribed today,
and p < 0.0001, respectively), however dose adjustments were with the remaining ~80% of prescriptions written by primary-care
P. Bradley et al. / Journal of Psychiatric Research 96 (2018) 100e107 105

Table 2
a Patients Achieving Remission Percent Change in HAM-A scores for Patients with Anxiety.
Severe Depression
40%
Patients with Depression (%)

35% Diagnosis Experimental Group Control Group


P
N % Change (SD) N % Change (SD)
30%
25%
Patients with Anxiety or Both Depression/Anxiety
4 week visit 177 34% (32) 175 27% (31) 0.05
20%
8 week visit 165 45% (33) 169 37% (33) 0.02
13%
12 week visit 159 51% (33) 162 44% (33) 0.06
9%
10% Patients with Anxiety Only
4 week visit 95 39% (32) 103 26% (31) 0.004
8 week visit 89 47% (32) 99 35% (35) 0.02
0% 12 week visit 82 54% (35) 95 42% (34) 0.02
8 weeks 12 weeks
Control NeuroIDgenetix Percent change in HAM-A scores is represented as mean change (±SD %). P values
were calculated using two sided t-test.

Patients Achieving Response Fig. 3. Response Rates for Patients with Anxiety. Response rates (50% reduction in
c Moderate/Severe Depression
HAM-A scores) in the experimental group (red bars, n ¼ 109) compared to the control
70% group (blue bars, n ¼ 115) for moderate and severe anxiety only patients. The response
Patients with Depression (%)

64%
60% rate by 12 weeks was higher in the experimental group (p ¼ 0.04). P value was
calculated using two-sided Fisher's exact test.
49% 46%
50%
41%
40%
30%
Table 3
20% Proportion of patients experiencing at least one medication change.
10% Medication Change Control Group Experimental P
0% (n ¼ 333) Group (n ¼ 352)
8 weeks 12 weeks
N % Changes N % Changes
Control NeuroIDgenetix Any Medication Change
2 weeks 212 64% 284 81%
4 weeks 105 32% 131 37%
Fig. 2. Remission and Response Rates for Patients with Depression. (a) Remission rates
8 weeks 75 23% 91 26%
(HAM-D17 scores 7) and (b) response rates (50% reduction in HAM-D17 scores) for
12 weeks 30 9% 46 13% <0.0001
the experimental group (red bars, n ¼ 40) compared to the control group (blue bars,
New Medication
n ¼ 53) for patients with severe depression. Percentage of patients achieving remission
2 weeks 151 45% 240 68%
and response at 12 weeks was higher in the experimental group (p ¼ 0.02, p ¼ 0.001
4 weeks 50 15% 62 18%
respectively). (c) Response rates for patients with moderate and severe depression
8 weeks 30 9% 31 9%
(HAM-D17 scores 20) were higher (p ¼ 0.01) in the experimental group (red bars,
12 weeks 4 1% 13 4% <0.0001
n ¼ 140) versus control (blue bars, n ¼ 121). P values were calculated using two-sided
Medication Discontinued
Fisher's exact test.
2 weeks 27 8% 91 26%
4 weeks 33 10% 44 13%
8 weeks 23 7% 28 8%
12 weeks 7 2% 22 6% <0.0001
physicians (Mark et al., 2009). Considering the majority of anxiety Dose Adjustment
and depression patients are being treated by primary care physi- 2 weeks 79 24% 70 20%
cians, pharmacogenetic tests which demonstrate utility in psychi- 4 weeks 56 17% 73 21%
8 weeks 46 14% 62 18%
atric and non-psychiatric practices are likely to enhance the overall 12 weeks 22 7% 25 7% 0.70
quality of psychiatric treatment. While previous studies have found
P values were calculated using two-sided Fisher's exact test.
clinical benefits of PGx-guided medication management in patients
with depression, the results are limited in their applicability to real-
world settings; excluding patients with comorbid psychiatric dis- pharmacogenetic testing in a manner that most accurately reflects
orders and enrolling patients only seen at psychiatric clinics. The clinical practice today. In order to achieve this, our study enrolled
aim of this study was to demonstrate the clinical utility of patients from 20 independent clinical sites throughout the US, with
106 P. Bradley et al. / Journal of Psychiatric Research 96 (2018) 100e107

treating physicians specializing in Psychiatry, Internal Medicine, NeuroIDgenetix® report) we believe this represents a clinical
Obstetrics and Gynecology, and Family Medicine. Patients enrolled “trade-off” between implementing a new treatment with an
included those new to therapy or those currently failing treatment. increased probability of effectiveness and the patient's awareness
Finally, our study population included patients with a broad range of new side effects associated with titrating medications. Given that
of severities for depression or anxiety, as well as comorbid patients medication changes were higher in the experimental group, and
displaying both depression and anxiety. ADE occurrence rates were statistically indistinguishable between
We observed statistically significant improvements for depres- the experimental and control groups, we believe the ADE
sion patients in the experimental group, in both the time to messaging in the IDgenetix® algorithm effectively provides a crit-
remission and response, as measured by HAM-D17. These results ical balance in improving efficacy, without a subsequent increase in
are well aligned with the growing body of scientific evidence the incidence or severity of ADEs.
demonstrating that pharmacogenetic information can improve This study demonstrates the clinical validity and utility of
antidepressant efficacy (Hall-Flavin et al., 2012, 2013; Winner et al., pharmacogenetic-guided treatment for depression and anxiety.
2013). In order to compare our findings with previously published While medication selection based on pharmacogenetic testing
studies, we focused a post-hoc analysis on the ‘Inadequately continues to become closer to routine clinical practice, test imple-
Controlled’ cohort, as published literature has focused exclusively mentation will result from pharmacogenetic algorithms that pro-
on the effects of pharmacogenetic-guided treatment in patients vide clinically validated information in an easily actionable format.
with treatment-resistant depression. An improvement in remission In alignment with these requirements, this study demonstrates
(42% vs. 27%, p-value ¼ 0.03) rates was evident in the experimental that the NeuroIDgenetix® test improves outcomes for patients with
group compared to control. Similarly, the experimental group also both depression and anxiety, in a diverse set of relevant clinical
showed greater improvement (62% vs. 44%, p-value ¼ 0.01) in settings.
response rates than the control arm. We believe this post-hoc
analysis demonstrates strong alignment with what has already Conflict of interest
been reported in the current literature. Furthermore, the design of
the study expands the utility of pharmacogenetic testing to include P. Bradley, M. Shiekh, V. Mehra, K. Vrbicky and S. Layle have no
‘New to Treatment’ patients as well as administration of the test in conflict of interest to disclose.
both psychiatric and non-psychiatric settings. M. Olson, A. Maciel, A. Cullors, J. Garces, and A. Lukowiak are
In addition, we believe this to be the first report of using phar- employed by AltheaDx.
macogenetics to improve outcomes for patients diagnosed with
anxiety. While statistical significance was found in the percent Acknowledgments
reduction of HAM-A scores of the experimental group comprised of
‘anxiety or both depression/anxiety’ subjects, ‘anxiety only’ sub- We thank the other investigators, clinical site personnel and the
jects appeared to benefit even more from pharmacogenetic- participants of this study for their valuable contributions. We thank
guidance. While additional work may be required to explain this Dr. Richard Weisler, Dr. Mustafa Hussain, Dr. Robert Detweiler and
difference, this outcome may reflect a unique set of pharmaco- Dr. Jeffrey Ross for their input on study design. We also thank Dr.
therapy requirements for treating anxiety in comorbid patients or it Ping-Yu Liu (Liu Associates) for statistical analysis and Innovis, LLC
may represent a clinical prioritization of managing depression for their assistance with clinical trial management. Finally, we
symptoms ahead of anxiety. thank Joel Centeno for his contributions throughout the entirety of
The blind nature of this clinical trial refers to blinding of both the project.
subjects and rating clinicians providing efficacy assessments. This research was funded by AltheaDx. The statistical analyses
Specimen collection occurred for all subjects regardless of study were conducted by an independent company, Liu Associates, and
arm to maintain blinding, however, patients were not asked to were paid for by AltheaDx.
guess their treatment assignment at the end of the study. While the
treating physicians could not be blinded to patient arm assignment, Appendix A. Supplementary data
they were blind to the HAM assessment scores, thus ensuring that
any expectancy bias did not affect the clinical outcomes measured. Supplementary data related to this article can be found at
Similarly, clinicians conducting the HAM assessments were blind to https://doi.org/10.1016/j.jpsychires.2017.09.024.
the therapeutic decisions made by the prescribing physician and
blind to patient treatment arm assignment. Additionally, all References
participating site staff (including physicians, study coordinators
and HAM-raters) were thoroughly trained on the protocol and Altar, C.A., Hornberger, J., Shewade, A., Cruz, V., Garrison, J., Mrazek, D., 2013.
Clinical validity of cytochrome P450 metabolism and serotonin gene variants in
followed procedures designed to maintain blinding. psychiatric pharmacotherapy. Int. Rev. Psychiatry 25, 509e533.
A critical attribute of any diagnostic test, and perhaps the most Anxiety and Depression Association of America, Updated August, 2016. Facts and
significant demonstration of clinical utility, is the extent to which a Statistics. https://www.adaa.org (Accessed on 8 February, 2017).
Barbui, C., Cipriani, A., Patel, V., Ayuso-Mateos, J.L., van Ommeren, M., 2011. Efficacy
test is implemented by a health care provider. In this study, phy- of antidepressants and benzodiazepines in minor depression: systematic re-
sicians made at least one medication change in 81% of subjects in view and meta-analysis. Br. J. Psychiatry 198 (1), 11e16.
the experimental group as compared to 64% of subjects in the Bystritsky, A., 2006. Treatment-resistant anxiety disorders. Mol. Psychiatry 11,
805e814.
control group (p < 0.0001) at the two week time-point. These re- Caudle, K.E., Klein, T.E., Hoffman, J.M., Muller, D.J., Whirl-Carillo, M., Gong, L., et al.,
sults strongly suggest that the NeuroIDgenetix® test results pro- 2014. Incorporation of pharmacogenomics into routine clinical practice: the
vided physicians with clear concise information to justify Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline
development process. Curr. Drug Metab. 15, 209e217.
additional, meaningful medication changes. While these additional,
Cusin, C., Yang, H., Yeung, A., Fava, M., 2010. Rating scales for depression. In: Baer, L.,
pharmacogenetically-guided changes are the likely cause of Blais, M.A. (Eds.), Handbook of Clinical Rating Scales and Assessment in Psy-
improved efficacy in treatment, no significant difference could be chiatry and Mental Health. Humana Press a Part of Springer Science, New York,
found between the experimental and control groups in regards to NY, pp. 7e35.
Evans, W.E., Johnson, J.A., 2001. Pharmacogenomics: the inherited basis for inter-
ADEs. While these results were initially surprising (as the avoid- individual differences in drug response. Annu. Rev. Genomics Hum. Genet. 2,
ance of ADEs is an integral part of the messaging in the 9e39.
P. Bradley et al. / Journal of Psychiatric Research 96 (2018) 100e107 107

Evans, W.E., Relling, M.V., 1999. Pharmacogenomics: translating functional geno- Mrazek, D.A., 2010. Psychiatric pharmacogenomic testing in clinical practice. Di-
mics into rational therapeutics. Science 286, 487e491. alogues Clin. Neurosci. 12, 69e76.
Fava, M., Rush, A.J., Alpert, J.E., Balasubramani, G.K., Wisniewski, S.R., Carmin, C.N., Porcelli, S., Drago, A., Fabbri, C., Gibiino, S., Calati, R., Serretti, A., 2011. Pharmaco-
et al., 2008. Difference in treatment outcome in outpatients with anxious versus genetics of antidepressant response. J. Psychiatry Neurosci. 36, 87e113. https://
nonanxious depression: a STAR*D report. Am. J. Psychiatry 165, 342e351. doi.org/10.1503/jpn.100059.
Gelenberg, A.J., Freeman, M.P., Markowitz, J.C., Rosenbaum, J.F., Thase, M.E., Pratt, L.A., Brody, D.J., 2014 Dec. Depression in the U.S. household population, 2009-
Trivedi, M.H., Oct. 2010. Practice Guideline for the Treatment of Patients with 2012. NCHS Data Brief. 172, 1e8.
Major Depressive Disorder. American Psychiatric Association. Available at: Pratt, V.M., Zehnbauer, B., Wilson, J.A., Baak, R., Babic, N., Bettinotti, M., et al., 2010.
https://psychiatryonline.org/pb/assets/raw/sitewide/practice_guidelines/ Characterization of 107 genomic DNA reference materials for CYP2D6, CYP2C19,
guidelines/mdd.pdf (Accessed on 10 February 2017). CYP2C9, VKORC1, and UGT1A1: a GeT-RM and Association for Molecular Pa-
Goodman, W.K., 2004. Selecting pharmacotherapy for generalized anxiety disorder. thology collaborative project. J. Mol. Diagn 12, 835e846.
J. Clin. Psychiatry 65 (13 Suppl. l), 8e13. Rush, A.J., Trivedi, M.H., Wisniewski, S.R., Nierenberg, A.A., Stewart, J.W., Warden, D.,
Greenberg, P.E., Fournier, A.A., Sisitsky, T., Pike, C.T., Kessler, R.C., 2015. The eco- et al., 2006. Acute and longer-term outcomes in depressed outpatients
nomic burden of adults with major depressive disorder in the United States requiring one or several treatment steps: a STAR*D report. Nov Am. J. Psychiatry
(2005 and 2010). J. Clin. Psychiatry 76, 155e162. 163, 1905e1917.
Hall-Flavin, D.K., Winner, J.G., Allen, J.D., Jordan, J.J., Nesheim, R.S., Snyder, K.A., Sheehan, D.V., Lecrubier, Y., Sheehan, K.H., Amorim, P., Janavs, J., Weiller, E., et al.,
et al., 2012. Using a pharmacogenomic algorithm to guide the treatment of 1998. The Mini-International Neuropsychiatric Interview (M.I.N.I.): the devel-
depression. Transl. Psychiatry 2, e172. https://doi.org/10.1038/tp.2012.99. opment and validation of a structured diagnostic psychiatric interview for
Hall-Flavin, D.K., Winner, J.G., Allen, J.D., Carhart, J.M., Proctor, B., Snyder, K.A., et al., DSM-IV and ICD-10. J. Clin. Psychiatry 59 (20 Suppl. l), 22e33.
2013. Utility of integrated pharmacogenomic testing to support the treatment Singh, A.B., 2015. Improved antidepressant remission in major depression via a
of major depressive disorder in a psychiatric outpatient setting. Pharmacogenet pharmacokinetic pathway polygene pharmacogenetic report. Clin. Psycho-
Genomics 23, 535e548. pharmacol. Neurosci. 13 (2), 150e156 [PMC free article] [PubMed] [Reference
Hamilton, M., 1959. The assessment of anxiety states by rating. Br. J. Med. Psychol. list].
32, 50e55. Siu, A.L., U.S. Preventative services Task Force, 2016. Screening for depression in
Hamilton, M., 1960. A rating scale for depression. J. Neurol. Neurosurg. Psychiatry adults US preventative services Task Force recommendation statement. JAMA
23, 56e62. 315, 380e387.
Helfand, M., Peterson, K., Dieperink, E., Ferguson, L., Anderson, J., May 2016. Evi- Trivedi, M.H., Rush, A.J., Wisniewski, S.R., Nierenberg, A.A., Warden, D., Ritz, L., et al.,
dence Brief: the Comparative Effectiveness, Harms and Cost-effectiveness of 2006. Evaluation of outcomes with citalopram for depression using
Pharmacogenomics-guided Antidepressant Treatment versus Usual Care for measurement-based care in STAR*D implications for clinical practice. Am. J.
Major Depressive Disorder. Evidence-based Synthesis Program Coordinating Psychiatry 163, 28e40.
Center, Portland CA Health Care System. U.S Department of Health and Human Services, Centers for Disease Control and
Hicks, J.K., Bishop, J.R., Sangkuhi, K., Muller, D.J., Ji, Y., Leckband, S.G., et al., 2015. Prevention, National Center for Health Statistics. Health, United States, 2013.
Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for With Special Feature on Prescription Drugs. Available at: https://www.cdc.gov/
CYP2D6 and CYP2C19 genotypes and dosing of selective serotonin reuptake nchs/data/hus/hus13.pdf (Accessed on 10, February 2017).
inhibitors. Clin. Pharmacol. Ther. 98, 127e134. U.S Food and Drug Administration. Table of pharmacogenomics biomarkers in drug
Huynh, N.N., McIntyre, R.S., 2008. What are the implications of the STAR*D Trial for labeling. www.fda.gov. Updated 7 November, 2016. (Accessed on 10 February,
primary care? A review and synthesis. Prim. Care Companion J. Clin. Psychiatry 2017).
10, 91e96. U.S. Food and Drug Administration, CDER, CBER, CDRH, January 2013. Guidance for
Kessler, R.C., Berglund, P., Demler, O., et al., 2005. Lifetime prevalence and age-of- Industry, Clinical Pharmacogenomics: Premarket Evaluation in Early-phase
onset distributions of DSM-IV disorders in the National Comorbidity Survey Clinical Studies and Recommendations for Labeling.
Replication. Arch. Gen. Psychiatry 62, 593e602. Whirl-Carrillo, M., McDonagh, E.M., Hebert, J.M., Gong, L., Sangkuhl, K., Thorn, C.F.,
Kessler, R.C., Birnbaum, H., Shahly, V., Bromet, E., Hwang, I., McLaughlin, K.A., 2010. Altman, R.B., Klein, T.E., 2012. Pharmacogenomics knowledge for personalized
Age differences in the prevalence and comorbidity of DSM-IV major depressive medicine. Clin. Pharmacol. Ther. 92 (4), 414e417. https://www.pharmgkb.org/
episodes: results from the WHO World Mental Health Survey Initiative. April page/pgxGeneRef.
Depress Anxiety 27 (4), 351e364. https://doi.org/10.1002/da.20634. Winner, J.G., Carhart, J.M., Altar, C.A., Allen, J.D., Dechairo, B.M., et al., 2013.
Lesch, K.-P., Bengel, D., Heils, A., Sabol, S.Z., Greenberg, B.D., Petri, S., et al., 1996. A Prospective, randomized, double-blind study assessing the clinical impact of
Association of anxiety-related traits with a polymorphism in the serotonin integrated pharmacogenomic testing for major depressive disorder. Discov.
transporter gene regulatory region. Science 274, 1527e1531. Med. 16, 219e227.
Mark, T.L., Levit, K.R., Buck, J.A., 2009. Psychotropic drug prescriptions by medical Wittchen, H.U., Kessler, R.C., Beesdo, K., Krause, P., Hofler, M., Hoyer, J., 2002.
specialty. Psychiatr. ServSept 60 (9), 1667. https://doi.org/10.1176/ps_ Generalized anxiety and depression in primary care: prevalence, recognition,
2009.60.9.1167. and management. J. Clin. Psychiatry 63, 24e34.

Você também pode gostar