Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
2.
Transl Psychiatry ; 12(1): 101, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35288545

ABSTRACT

The pharmacological treatment of depression consists of stages of trial and error, with less than 40% of patients achieving remission during first medication trial. However, in a large, randomized-controlled trial (RCT) in the U.S. ("GUIDED"), significant improvements in response and remission rates were observed in patients who received treatment guided by combinatorial pharmacogenomic testing, compared to treatment-as-usual (TAU). Here we present results from the Canadian "GAPP-MDD" RCT. This 52-week, 3-arm, multi-center, participant- and rater-blinded RCT evaluated clinical outcomes among patients with depression whose treatment was guided by combinatorial pharmacogenomic testing compared to TAU. The primary outcome was symptom improvement (change in 17-item Hamilton Depression Rating Scale, HAM-D17) at week 8. Secondary outcomes included response (≥50% decrease in HAM-D17) and remission (HAM-D17 ≤ 7) at week 8. Numerically, patients in the guided-care arm had greater symptom improvement (27.6% versus 22.7%), response (30.3% versus 22.7%), and remission rates (15.7% versus 8.3%) compared to TAU, although these differences were not statistically significant. Given that the GAPP-MDD trial was ultimately underpowered to detect statistically significant differences in patient outcomes, it was assessed in parallel with the larger GUIDED RCT. We observed that relative improvements in response and remission rates were consistent between the GAPP-MDD (33.0% response, 89.0% remission) and GUIDED (31.0% response, 51.0% remission) trials. Together with GUIDED, the results from the GAPP-MDD trial indicate that combinatorial pharmacogenomic testing can be an effective tool to help guide depression treatment in the context of the Canadian healthcare setting (ClinicalTrials.gov NCT02466477).


Subject(s)
Depressive Disorder, Major , Pharmacogenomic Testing , Antidepressive Agents/therapeutic use , Canada , Depression , Depressive Disorder, Major/chemically induced , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Humans , Treatment Outcome
4.
Am J Geriatr Psychiatry ; 28(9): 933-945, 2020 09.
Article in English | MEDLINE | ID: mdl-32513518

ABSTRACT

OBJECTIVE: Evaluate the clinical utility of combinatorial pharmacogenomic testing for informing medication selection among older adults who have experienced antidepressant medication failure for major depressive disorder (MDD). DESIGN: Post hoc analysis of data from a blinded, randomized controlled trial comparing two active treatment arms. SETTING: Psychiatry specialty and primary care clinics across 60 U.S. community and academic sites. PARTICIPANTS: Adults age 65 years or older at baseline (n = 206), diagnosed with MDD and inadequate response to at least one medication on the combinatorial pharmacogenomic test report during the current depressive episode. INTERVENTION: Combinatorial pharmacogenomic testing to inform medication selection (guided-care), compared with treatment as usual (TAU). OUTCOMES: Mean percent symptom improvement, response rate, and remission rateat week 8, measured using the 17-item Hamilton Depression Rating Scale; medication switching; and comorbidity moderator analysis. RESULTS: At week 8, symptom improvement was not significantly different for guided-care than for TAU (∆ = 8.1%, t = 1.64, df = 187; p = 0.102); however, guided-care showed significantly improved response (∆ = 13.6%, t = 2.16, df = 187; p = 0.032) and remission (∆ = 12.7%, t = 2.49, df = 189; p = 0.014) relative to TAU. By week 8, more than twice as many patients in guided-care than in TAU were on medications predicted to have no gene-drug interactions (χ2 = 19.3, df = 2; p <0.001). Outcomes in the guided-care arm showed consistent improvement through the end of the open-design 24-week trial, indicating durability of the effect. Differences in outcomes between arms were not significantly impacted by comorbidities. CONCLUSIONS: Combinatorial pharmacogenomic test-informed medication selection improved outcomes over TAU among older adults with depression.


Subject(s)
Antidepressive Agents , Depressive Disorder, Major , Pharmacogenomic Testing/methods , Aged , Antidepressive Agents/administration & dosage , Antidepressive Agents/classification , Antidepressive Agents/pharmacokinetics , Depressive Disorder, Major/diagnosis , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Drug Substitution , Female , Humans , Male , Outcome Assessment, Health Care , Patient Selection , Psychiatric Status Rating Scales , Treatment Failure
5.
Pharmacogenomics ; 21(8): 521-531, 2020 06.
Article in English | MEDLINE | ID: mdl-32301648

ABSTRACT

Aim: Evaluate the cost-effectiveness of combinatorial pharmacogenomic (PGx) testing, versus treatment as usual (TAU), to guide treatment for patients with depression, from the Canadian public healthcare system perspective. Materials & methods: Clinical and economic data associated with depression were extracted from published literature. Clinical (quality-adjusted life years; QALYs) and economic (incremental cost-effectiveness ratio) outcomes were modeled using combinatorial PGx and TAU treatment strategies across a 5-year time horizon. Results: With the combinatorial PGx strategy to guide treatment, patients were projected to gain 0.14-0.19 QALYs versus TAU. Accounting for test price, combinatorial PGx saved CAD $1,687-$3,056 versus TAU. Incremental cost-effectiveness ratios ranged from -$11,861 to -$16,124/QALY gained. Conclusion: Combinatorial PGx testing was more efficacious and less costly compared with the TAU for depression.


Subject(s)
Cost-Benefit Analysis/methods , Depression/economics , Depression/epidemiology , National Health Programs/economics , Pharmacogenomic Testing/economics , Pharmacogenomic Testing/methods , Canada/epidemiology , Depression/diagnosis , Humans
6.
Pharmacogenomics ; 21(8): 559-569, 2020 06.
Article in English | MEDLINE | ID: mdl-32301649

ABSTRACT

Aim: To perform a meta-analysis of prospective, two-arm studies examining the clinical utility of using the combinatorial pharmacogenomic test, GeneSight Psychotropic, to inform treatment decisions for patients with major depressive disorder (MDD). Patients & methods: The pooled mean effect of symptom improvement and pooled relative risk ratio (RR) of response and remission were calculated using a random effect model. Results: Overall, 1556 patients were included from four studies, with outcomes evaluated at week 8 or week 10. Patient outcomes were significantly improved for patients with MDD whose care was guided by the combinatorial pharmacogenomic test results compared with unguided care (symptom improvement Δ = 10.08%, 95% CI: 1.67-18.50; p = 0.019; response RR = 1.40, 95% CI: 1.17-1.67; p < 0.001; remission RR = 1.49, 95% CI: 1.17-1.89; p = 0.001). Conclusion: GeneSight Psychotropic guided care improves outcomes among patients with MDD.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Pharmacogenomic Testing/methods , Antidepressive Agents/adverse effects , Depressive Disorder, Major/epidemiology , Humans , Prospective Studies , Psychotropic Drugs/adverse effects , Psychotropic Drugs/therapeutic use
7.
J Geriatr Psychiatry Neurol ; 33(6): 324-332, 2020 11.
Article in English | MEDLINE | ID: mdl-31842673

ABSTRACT

OBJECTIVE: We compared economic outcomes when elderly patients with neuropsychiatric disorders received psychotropic medications guided by a combinatorial pharmacogenomic (PGx) test. METHODS: This is a subanalysis of a 1-year prospective assessment of medication cost for patients with neuropsychiatric disorders receiving combinatorial PGx testing. Pharmacy claims were used to compare per member per year (PMPY) medication cost for patients ≥65 and <65 years old when medications were congruent or incongruent with the PGx test. Polypharmacy was also assessed. RESULTS: Congruent prescribing was associated with savings of US$3497 PMPY (P < .001) for patients ≥65 years and US$2467 PMPY (P < .001) for patients <65, compared to incongruent prescribing. Congruent prescribing in patients ≥65 treated by primary care providers was associated with US$4113 PMPY (P = .026) in savings, while congruent prescribing by psychiatrists was associated with US$120 PMPY (P = .719). Congruent prescribing was also associated with one fewer neuropsychiatric medication for patients ≥65 (P = .070). CONCLUSION: Congruence with PGx testing was associated with medication cost savings in elderly patients.


Subject(s)
Drug Prescriptions/statistics & numerical data , Genetic Testing/economics , Mental Disorders/drug therapy , Pharmacogenetics/economics , Pharmacogenomic Testing/economics , Psychotropic Drugs/economics , Aged , Antidepressive Agents/economics , Antidepressive Agents/therapeutic use , Antipsychotic Agents/economics , Antipsychotic Agents/therapeutic use , Drug Costs/statistics & numerical data , Fees, Pharmaceutical/statistics & numerical data , Female , Genetic Testing/methods , Geriatric Psychiatry , Humans , Male , Mental Disorders/psychology , Middle Aged , Pharmacogenetics/methods , Prescription Drugs/economics , Prospective Studies , Psychotropic Drugs/therapeutic use
8.
Can J Psychiatry ; 65(5): 338-346, 2020 05.
Article in English | MEDLINE | ID: mdl-31835904

ABSTRACT

OBJECTIVES: To characterize the health-care utilization and economic burden associated with depression in Manitoba, Canada. METHODS: Patient-level data were retrieved from the Manitoba Centre for Health Policy administrative, clinical, and laboratory databases for the study period of January 1, 1996, through December 31, 2016. Patients were assigned to the depression cohort based on diagnoses recorded in hospitalizations and outpatient physician claims, as well as antidepressant prescription drug claims. A comparison cohort of nondepressed subjects, matched with replacement for age, gender, place of residence (urban vs. rural), and index date, was created. Demographics, comorbidities, intentional self-harm, mortality, health-care utilization, prescription drug utilization, and costs of health-care utilization and social services were compared between depressed patients and matched nondepressed patients, and incidence rate ratios and hazard ratios were reported. RESULTS: There were 190,065 patients in the depression cohort and 378,177 patients in the nondepression cohort. Comorbidities were 43% more prevalent among depressed patients. Intentional self-harm, all-cause mortality, and suicide mortality were higher among patients with depression than the nondepression cohort. Health-care utilization-including hospitalizations, physician visits, physician-provided psychotherapy, and prescription drugs-was higher in the depression than the nondepression cohort. Mean health-care utilization costs were 3.5 times higher among depressed patients than nondepressed patients ($10,064 and $2,832, respectively). Similarly, mean social services costs were 3 times higher ($1,522 and $510, respectively). Overall, depression adds a total average cost of $8,244 (SD = $40,542) per person per year. CONCLUSIONS: Depression contributes significantly to health burden and per patient costs in Manitoba, Canada. Extrapolation of the results to the entire Canadian health-care system projects an excess of $12 billion annually in health system spending.


Subject(s)
Cost of Illness , Depression , Canada , Depression/epidemiology , Health Care Costs , Humans , Manitoba/epidemiology , Retrospective Studies
9.
Clinicoecon Outcomes Res ; 11: 779-787, 2019.
Article in English | MEDLINE | ID: mdl-31849503

ABSTRACT

OBJECTIVE: To estimate Canadian pharmacy cost savings associated with psychiatric medication prescribing that is guided by combinatorial pharmacogenomic testing in patients switching or augmenting their psychiatric medication. METHODS: Pharmacy claims data from a United States (US) pharmacy benefit manager were analyzed for 1662 patients who recently augmented or switched to a different antidepressant or antipsychotic medication and underwent combinatorial pharmacogenomic testing. Costs of prescription medications were translated to the Canadian healthcare system by matching drug names and doses using the Ontario Drug Benefit Formulary. One-year costs (2017 CAD) were compared between patients whose clinician prescribed antidepressants or antipsychotics that were consistent (congruent) or inconsistent (incongruent) with the combinatorial pharmacogenomic test recommendations. RESULTS: Patients whose psychiatric medication treatment was congruent with the combinatorial pharmacogenomic test report saved $1061 CAD per member per year (PMPY) on prescription medication costs relative to patients whose medications were incongruent with their test report (p<0.0001). For patients ages <65 and ≥65, prescription medication costs were $979 and $1178 CAD PMPY lower, respectively, for patients who followed the report recommendations (p=0.0004 and p=0.13). Prescription drug fills from the US pharmacy claims were concordant with the Canadian Formulary; 62% of fills matched at both the drug name and dose strength, 81% matched at drug name, and >99% matched at the therapeutic chapter. CONCLUSIONS: Antidepressant and antipsychotic prescribing that was congruent with combinatorial pharmacogenomic test guidance was associated with significant cost savings on Canadian prescription medications according to the Ontario Drug Benefit Formulary.

11.
J Psychiatr Res ; 111: 59-67, 2019 04.
Article in English | MEDLINE | ID: mdl-30677646

ABSTRACT

Current prescribing practices for major depressive disorder (MDD) produce limited treatment success. Although pharmacogenomics may improve outcomes by identifying genetically inappropriate medications, studies to date were limited in scope. Outpatients (N = 1167) diagnosed with MDD and with a patient- or clinician-reported inadequate response to at least one antidepressant were enrolled in the Genomics Used to Improve DEpression Decisions (GUIDED) trial - a rater- and patient-blind randomized controlled trial. Patients were randomized to treatment as usual (TAU) or a pharmacogenomics-guided intervention arm in which clinicians had access to a pharmacogenomic test report to inform medication selections (guided-care). Medications were considered congruent ('use as directed' or 'use with caution' test categories) or incongruent ('use with increased caution and with more frequent monitoring' test category) with test results. Unblinding occurred after week 8. Primary outcome was symptom improvement [change in 17-item Hamilton Depression Rating Scale (HAM-D17)] at week 8; secondary outcomes were response (≥50% decrease in HAM-D17) and remission (HAM-D17 ≤ 7) at week 8. At week 8, symptom improvement for guided-care was not significantly different than TAU (27.2% versus 24.4%, p = 0.107); however, improvements in response (26.0% versus 19.9%, p = 0.013) and remission (15.3% versus 10.1%, p = 0.007) were statistically significant. Patients taking incongruent medications prior to baseline who switched to congruent medications by week 8 experienced greater symptom improvement (33.5% versus 21.1%, p = 0.002), response (28.5% versus 16.7%, p = 0.036), and remission (21.5% versus 8.5%, p = 0.007) compared to those remaining incongruent. Pharmacogenomic testing did not significantly improve mean symptoms but did significantly improve response and remission rates for difficult-to-treat depression patients over standard of care (ClinicalTrials.gov NCT02109939).


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Major/metabolism , Outcome Assessment, Health Care , Pharmacogenomic Testing , Adolescent , Adult , Aged , Aged, 80 and over , Antidepressive Agents/administration & dosage , Antidepressive Agents/adverse effects , Cohort Studies , Cytochrome P-450 Enzyme System/genetics , Double-Blind Method , Female , Humans , Male , Middle Aged , Receptor, Serotonin, 5-HT2A/genetics , Remission Induction , Serotonin Plasma Membrane Transport Proteins/genetics , Selective Serotonin Reuptake Inhibitors/pharmacology , Young Adult
13.
J Psychiatr Res ; 104: 157-162, 2018 09.
Article in English | MEDLINE | ID: mdl-30081389

ABSTRACT

Failed medication trials are common in the treatment of major depressive disorder (MDD); however, the use of combinatorial pharmacogenomics to guide medication selection has been previously associated with improved outcomes in the psychiatric care setting. The utility of combinatorial pharmacogenomics in patients with MDD in primary care and psychiatric care settings was evaluated here. Patients enrolled in a naturalistic, open-label, prospective study [Individualized Medicine: Pharmacogenetics Assessment and Clinical Treatment (IMPACT)] with MDD were evaluated (N = 1871). Pharmacogenomic testing was performed for all patients and medications were categorized based on gene-drug interactions. Beck's Depression Inventory (BDI) was evaluated at baseline and follow-up (weeks 8-12). Symptom improvement (percent decrease in BDI), response (≥50% decrease in BDI), and remission (BDI≤10) at follow-up were evaluated according to provider type and whether medications were genetically congruent (little/no gene-drug interactions). There was a 27.9% reduction in depression symptoms at follow-up, as well as response and remission rates of 25.7% and 15.2%, respectively. Outcomes were significantly better among patients treated by primary care providers versus psychiatrists (symptom improvement 31.7% versus 24.9%, p < 0.01; response rate 30.1% versus 22.3%, p < 0.01; remission rate 19.5% versus 12.0%, p < 0.01). There was a 31% relative improvement in response rate among patients taking congruent versus incongruent medications, with slightly higher congruence among primary care providers (87.6%) versus psychiatrists (85.2%). Following combinatorial pharmacogenomic testing, outcomes were significantly improved among patients treated by primary care providers compared to psychiatrists, which supports the use of pharmacogenomics in broader treatment settings.


Subject(s)
Depressive Disorder, Major/genetics , Depressive Disorder, Major/therapy , Pharmacogenetics , Physicians, Primary Care , Psychiatry , Treatment Outcome , Adult , Antidepressive Agents/therapeutic use , Female , Humans , Male , Middle Aged , Prospective Studies , Psychiatric Status Rating Scales
14.
Per Med ; 15(3): 189-197, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29739269

ABSTRACT

AIM: The aim of this study was to validate the analytical performance of a combinatorial pharmacogenomics test designed to aid in the appropriate medication selection for neuropsychiatric conditions. MATERIALS & METHODS: Genomic DNA was isolated from buccal swabs. Twelve genes (65 variants/alleles) associated with psychotropic medication metabolism, side effects, and mechanisms of actions were evaluated by bead array, MALDI-TOF mass spectrometry, and/or capillary electrophoresis methods (GeneSight Psychotropic, Assurex Health, Inc.). RESULTS: The combinatorial pharmacogenomics test has a dynamic range of 2.5-20 ng/µl of input genomic DNA, with comparable performance for all assays included in the test. Both the precision and accuracy of the test were >99.9%, with individual gene components between 99.4 and 100%. CONCLUSION: This study demonstrates that the combinatorial pharmacogenomics test is robust and reproducible, making it suitable for clinical use.


Subject(s)
Mental Disorders/genetics , Pharmacogenomic Testing/methods , Psychotropic Drugs/pharmacokinetics , Algorithms , DNA/analysis , Gene Frequency , Humans , Mental Disorders/drug therapy , Pharmacogenomic Variants
15.
Am J Drug Alcohol Abuse ; 44(4): 431-440, 2018.
Article in English | MEDLINE | ID: mdl-29333880

ABSTRACT

BACKGROUND: Currently, no pharmacogenetic tests for selecting an opioid-dependence pharmacotherapy have been approved by the US Food and Drug Administration. OBJECTIVES: Determine the effects of variants in 11 genes on dropout rate and dose in patients receiving methadone or buprenorphine/naloxone (ClinicalTrials.gov Identifier: NCT00315341). METHODS: Variants in six pharmacokinetic genes (CYP1A2, CYP2B6, CYP2C19, CYP2C9, CYP2D6, CYP3A4) and five pharmacodynamic genes (HTR2A, OPRM1, ADRA2A, COMT, SLC6A4) were genotyped in samples from a 24-week, randomized, open-label trial of methadone and buprenorphine/naloxone for the treatment of opioid dependence (n = 764; 68.7% male). Genotypes were then used to determine the metabolism phenotype for each pharmacokinetic gene. Phenotypes or genotypes for each gene were analyzed for association with dropout rate and mean dose. RESULTS: Genotype for 5-HTTLPR in the SLC6A4 gene was nominally associated with dropout rate when the methadone and buprenorphine/naloxone groups were combined. When the most significant variants associated with dropout rate were analyzed using pairwise analyses, SLC6A4 (5-HTTLPR) and COMT (Val158Met; rs4860) had nominally significant associations with dropout rate in methadone patients. None of the genes analyzed in the study was associated with mean dose of methadone or buprenorphine/naloxone. CONCLUSIONS: This study suggests that functional polymorphisms related to synaptic dopamine or serotonin levels may predict dropout rates during methadone treatment. Patients with the S/S genotype at 5-HTTLPR in SLC6A4 or the Val/Val genotype at Val158Met in COMT may require additional treatment to improve their chances of completing addiction treatment. Replication in other methadone patient populations will be necessary to ensure the validity of these findings.


Subject(s)
Buprenorphine, Naloxone Drug Combination/therapeutic use , Genotype , Methadone/therapeutic use , Opioid-Related Disorders/drug therapy , Opioid-Related Disorders/genetics , Patient Dropouts , Adult , Female , Humans , Male , Narcotic Antagonists/therapeutic use , Opiate Substitution Treatment , Pharmacogenetics , Pharmacogenomic Testing , Treatment Outcome
16.
Clin Ther ; 39(3): 592-602.e1, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28238356

ABSTRACT

PURPOSE: This study was an analysis based on a previously completed prospective study investigating medication costs of patients with mental illness guided by using the GeneSight proprietary combinatorial pharmacogenomic (PGx) test. The primary objective of this study was to determine potential cost savings of combinatorial PGx testing over the course of 1 year in patients with mental illness treated by primary care providers (PCPs) and psychiatrists who had switched or added a new psychiatric medication after patients failed to respond to monotherapy. The current evaluation details cost savings of treatment decisions congruent and incongruent with the combinatorial PGx test recommendations specific to PCPs and psychiatrists. METHODS: This study was a subanalysis of a 1-year, prospective trial comparing medication costs of 2168 patients undergoing GeneSight testing. Pharmacy claims were provided by a pharmacy benefits manager, comparing medication costs 6 months before combinatorial PGx testing and followed up for 1 year after the testing. This analysis compared congruence and cost savings per patient based on the type of health care provider administering care. FINDINGS: Using data from a large pharmacy benefits manager, we found that PCPs treat the majority of mental health patients receiving psychotropic medication prescriptions, including treatment-resistant patients. PCPs congruent with combinatorial PGx testing provided the most medication cost savings for payers and patients at $3988 per member per year (P < 0.001). IMPLICATIONS: Health care providers treating patients with mental illness can significantly reduce medication costs by following the combinatorial PGx report recommendations. PCPs, who treat the majority of patients with mental illness, reported a significant reduction in medication costs for both central nervous system and non-central nervous system drugs.


Subject(s)
Mental Disorders/drug therapy , Pharmacogenetics , Psychotropic Drugs/therapeutic use , Cost Savings , Drug Costs , Drug Prescriptions , Female , Humans , Male , Mental Health , Middle Aged , Pharmaceutical Services , Primary Health Care , Prospective Studies
17.
Yale J Biol Med ; 88(4): 375-82, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26604861

ABSTRACT

Pharmacogenomic testing in mental health has not yet reached its full potential. An important reason for this involves differentiating individual gene testing (IGT) from a combinatorial pharmacogenomic (CPGx) approach. With IGT, any given gene reveals specific information that may, in turn, pertain to a smaller number of medications. CPGx approaches attempt to encompass more complete genomic information by combining moderate risk alleles and synergistically viewing the results from the perspective of the medication. This manuscript will discuss IGT and CPGx approaches to psychiatric pharmacogenomics and review the clinical validity, clinical utility, and economic parameters of both.


Subject(s)
Genetic Testing , Mental Disorders/genetics , Genes/genetics , Genetic Testing/methods , Humans , Pharmacogenetics/methods
18.
Curr Med Res Opin ; 31(9): 1633-43, 2015.
Article in English | MEDLINE | ID: mdl-26086890

ABSTRACT

OBJECTIVES: The objective of this project was to determine pharmacy cost savings and improvement in adherence based on a combinatorial pharmacogenomic test (CPGx ) in patients who had switched or added a new psychiatric medication after having failed monotherapy for their psychiatric disorder. RESEARCH DESIGN AND METHODS: The prospective project compared 1 year pharmacy claims between a GeneSight CPGx guided cohort and a propensity-matched control group. Patients were project eligible if they augmented or switched to a different antidepressant or antipsychotic medication within the previous 90 days. Following the medication switch or augmentation, pharmacogenomic (PGx) testing was offered to each patient's treating clinician. Pharmacy claims were extracted from the Medco pharmacy claims database for each patient (n = 2168) for 1 year following testing and compared to a 5-to-1 propensity-matched treatment as usual (TAU), standard of care control group (n = 10,880). MAIN OUTCOME MEASURES: Total pharmacy spend per member per year; adherence. RESULTS: Patients who received PGx testing saved $1035.60 in total medication costs (both CNS and non-CNS medications) over 1 year compared to the non-tested standard of care cohort (p = 0.007). PGx testing improved adherence compared to standard of care (ΔPDCCPGx = 0.11 vs ΔPDCTAU = -0.01; p < 0.0001). Pharmacy cost savings averaged $2774.53 for patients who were changed to a CPGx congruent medication regimen, compared to those who were not (p < 0.0001). CONCLUSIONS: PGx testing provides significant 'real world' cost savings, while simultaneously improving adherence in a difficult to treat psychiatric population. Limitations of this study include the lack of therapeutic efficacy follow-up data and possible confounding due to matching only on demographic and psychiatric variables.


Subject(s)
Antidepressive Agents , Antipsychotic Agents , Genetic Testing/economics , Pharmacogenetics , Adult , Aged , Antidepressive Agents/economics , Antidepressive Agents/therapeutic use , Antipsychotic Agents/economics , Antipsychotic Agents/therapeutic use , Cost Savings/statistics & numerical data , Drug Costs/statistics & numerical data , Fees, Pharmaceutical/statistics & numerical data , Female , Genetic Testing/methods , Humans , Male , Medication Adherence/psychology , Medication Adherence/statistics & numerical data , Mental Disorders/drug therapy , Mental Disorders/psychology , Middle Aged , Outcome Assessment, Health Care , Pharmacogenetics/economics , Pharmacogenetics/methods , Prospective Studies , United States
19.
Mol Neuropsychiatry ; 1(3): 145-55, 2015 Oct.
Article in English | MEDLINE | ID: mdl-27606312

ABSTRACT

DNA of 258 patients with treatment-resistant depression was collected in three 8-10 week, two-arm, prospective clinical trials. Forty-four allelic variations were measured in genes for the cytochrome P450 (CYP) enzymes CYP2D6, CYPC19, and CYP1A2, the serotonin transporter (SLC6A4), and the 5-HT2A receptor (HTR2A). The combinatorial pharmacogenomic (CPGx™) GeneSight test results were provided to clinicians to support medication changes from baseline (guided arm), or they were provided at the end of each study to clinicians of unguided patients who were treated as usual (TAU). TAU subjects who at baseline were prescribed medications genetically discordant for them showed only a 12% symptom improvement, far less than the 32.5% or 28.5% improvements of the TAU subjects on yellow-category ('use with caution'; p = 0.002) or green-category medications ('use as recommended'; p = 0.02), respectively. The odds of a clinical response were increased 2.3-fold among all GeneSight-guided compared to all TAU subjects (p = 0.004), and overall, the guided group had a 53% greater improvement in depressive symptoms (p = 0.0002), a 1.7-fold relative improvement in response (p = 0.01), and a number needed to treat for one clinical response above that seen in the TAU group of 6.07.

20.
Glob Adv Health Med ; 3(3): 25-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24891991

ABSTRACT

PRIMARY STUDY OBJECTIVE: To evaluate the economic utility of a fecal biomarker panel structured to suggest alternative, treatable diagnoses in patients with symptoms of irritable bowel syndrome (IBS) by quantifying, comparing, and contrasting health service costs between tested and non-tested patients. STUDY DESIGN: Retrospective, matched cohort study comparing direct medical costs for IBS patients undergoing fecal biomarker testing with those of matched control subjects. METHODS: We examined de-identified medical and pharmacy claims of a large American pharmacy benefit manager to identify plan members who underwent panel testing, were eligible for covered benefits for at least 180 days prior to the test date, and had data available for 30, 90, and 365 days after that date. We used propensity score matching to develop population-based control cohorts for each tested cohort, comprised of records with IBS-related diagnoses but for which panel testing was not performed. Primary outcome measures were diagnostic and medical services costs as determined from claims data. RESULTS: Two hundred nine records from tested subjects met inclusion criteria. The only significant baseline differences between groups were laboratory costs, which were significantly higher in each tested cohort. At each follow-up time point, total medical and gastrointestinal procedural costs were significantly higher in non-tested cohorts. Within tested cohorts, costs declined significantly from baseline, while costs rose significantly in non-tested control cohorts; these differences were also significant between groups at each time point. CONCLUSIONS: Structured fecal biomarker panel testing was associated with significantly lower medical and gastrointestinal procedural costs in this study of patients with IBS symptoms.

SELECTION OF CITATIONS
SEARCH DETAIL
...