Clinical utility of combinatorial pharmacogenomic testing in depression: A Canadian patient- and rater-blinded, randomized, controlled trial

Arun K Tiwari, Clement C Zai, C Anthony Altar, Julie-Anne Tanner, Paige E Davies, Paul Traxler, James Li, Elizabeth S Cogan, Matthew T Kucera, Ana Gugila, Nicole Braganza, Heather Emmerson, Gwyneth Zai, Daniel J Müller, Robert Levitan, Stefan Kloiber, Zafiris J Daskalakis, Benicio N Frey, James M Bowen, Jean-Eric Tarride, Richard Tytus, Ranjith Chandrasena, Nicholas Voudouris, Valerie H Taylor, Raymond Tempier, Verinder Sharma, Akshya Vasudev, Peter Dzongowski, Lew Pliamm, Todd Greenspoon, Bryan M Dechairo, James L Kennedy, Arun K Tiwari, Clement C Zai, C Anthony Altar, Julie-Anne Tanner, Paige E Davies, Paul Traxler, James Li, Elizabeth S Cogan, Matthew T Kucera, Ana Gugila, Nicole Braganza, Heather Emmerson, Gwyneth Zai, Daniel J Müller, Robert Levitan, Stefan Kloiber, Zafiris J Daskalakis, Benicio N Frey, James M Bowen, Jean-Eric Tarride, Richard Tytus, Ranjith Chandrasena, Nicholas Voudouris, Valerie H Taylor, Raymond Tempier, Verinder Sharma, Akshya Vasudev, Peter Dzongowski, Lew Pliamm, Todd Greenspoon, Bryan M Dechairo, James L Kennedy

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).

Conflict of interest statement

AKT is a co-inventor on a patent for antipsychotic-induced weight gain at the time of this study (U.S. patent no. 10,662,475). CCZ had a patent on antipsychotic-induced weight gain markers at the time of this study. CAA had ownership/partnership in Splice Therapeutics at the time of this study, and also serves on the advisory board for AxoSim Inc. and the board of directors for ASENT. JAT, PED, PT, JL, MTK, and AG were employed by Myriad Neuroscience/Assurex Health at the time of this study. MTK and ESC were employed by Myriad Genetics at the time of this study. DJM was a co-inventor on two patents assessing risk for antipsychotic-induced weight gain at the time of this study and was also a co-investigator on two pharmacogenetic studies where genetic test kits were provided as in-kind contribution by Myriad Neuroscience but did not receive any salary, equity, stocks, or options from any pharmacogenetic companies. SK received research support from the University of Toronto, Department of Psychiatry Academic Scholar Award, the Labatt Family Innovation Fund in Brain Health, and the Canadian Institutes of Health Research (CIHR) at the time of this study, and received honorarium from Empowerpharm for past consultation. JMB and JET received funding and payment from Myriad Neuroscience to conduct the GAPP-MDD trial until the contract was terminated by Myriad Neuroscience. BMD was employed by Myriad Genetics at the time of this study and received salary and stock options. JLK is an unpaid member of the Myriad Neuroscience Scientific Advisory Board. All other authors declare no conflicts of interest.

© 2022. The Author(s).

Figures

Fig. 1. HAM-D17 clinical outcomes at 8…
Fig. 1. HAM-D17 clinical outcomes at 8 weeks by treatment arm in the GAPP-MDD clinical trial and the previous GUIDED trial [14].
A Per-protocol (PP) cohort. GAPP-MDD trial: Guided care arm N = 127, TAU arm N = 69; GUIDED trial: Guided care arm N = 560, TAU arm N = 607. B Intent-to-treat (ITT) cohort. GAPP-MDD trial: Guided care arm N = 211, TAU arm N = 97); GUIDED trial: Guided care arm N = 621, TAU arm N = 677.
Fig. 2. Medication congruency by week in…
Fig. 2. Medication congruency by week in the Per-Protocol cohort of the GAPP-MDD clinical trial and the previous GUIDED trial [14].
The proportion of patients taking genetically congruent medications from the GAPP-MDD and GUIDED trials are shown in blue and orange, respectively. Squares represent Guided-Care treatment arms, and circles represent TAU treatment arms.
Fig. 3. Meta-analysis of randomized-controlled trials evaluating…
Fig. 3. Meta-analysis of randomized-controlled trials evaluating combinatorial pharmacogenomic testing.
A Meta-analysis of symptom improvement. B Meta-analysis of response. C Meta-analysis of remission.

References

    1. Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry. 2006;163:1905–17. doi: 10.1176/ajp.2006.163.11.1905.
    1. Hicks JK, Bishop JR, Sangkuhl K, Muller DJ, Ji Y, Leckband SG, et al. Clinical pharmacogenetics implementation consortium (CPIC) guideline for CYP2D6 and CYP2C19 genotypes and dosing of selective serotonin reuptake inhibitors. Clin Pharmacol Therapeutics. 2015;98:127–34. doi: 10.1002/cpt.147.
    1. Hicks JK, Sangkuhl K, Swen JJ, Ellingrod VL, Muller DJ, Shimoda K, et al. Clinical pharmacogenetics implementation consortium guideline (CPIC) for CYP2D6 and CYP2C19 genotypes and dosing of tricyclic antidepressants: 2016 update. Clin Pharm Ther. 2017;102:37–44. doi: 10.1002/cpt.597.
    1. KNNMP. Pharmacogenetics. 2021; .
    1. U.S. Food & Drug Administration. Table of Pharmacogenomic Biomarkers in Drug Labeling. 2020; .
    1. Bousman CA, Bengesser SA, Aitchison KJ, Amare AT, Aschauer H, Baune BT, et al. Review and consensus on pharmacogenomic testing in psychiatry. Pharmacopsychiatry. 2021;54:5–17.
    1. Bousman CA, Arandjelovic K, Mancuso SG, Eyre HA, Dunlop BW. Pharmacogenetic tests and depressive symptom remission: a meta-analysis of randomized controlled trials. Pharmacogenomics. 2019;20:37–47. doi: 10.2217/pgs-2018-0142.
    1. Rosenblat JD, Lee Y, McIntyre RS. The effect of pharmacogenomic testing on response and remission rates in the acute treatment of major depressive disorder: A meta-analysis. J Affect Disord. 2018;241:484–91. doi: 10.1016/j.jad.2018.08.056.
    1. Brown L, Vranjkovic O, Li J, Yu K, Al Habbab T, Johnson H, et al. The clinical utility of combinatorial pharmacogenomic testing for patients with depression: a meta-analysis. Pharmacogenomics. 2020;21:559–69. doi: 10.2217/pgs-2019-0157.
    1. Hall-Flavin DK, Winner JG, Allen JD, Jordan JJ, Nesheim RS, Snyder KA, et al. Using a pharmacogenomic algorithm to guide the treatment of depression. Transl Psychiatry. 2012;2:e172. doi: 10.1038/tp.2012.99.
    1. Altar CA, Carhart JM, Allen JD, Hall-Flavin DK, Dechairo BM, Winner JG. Clinical validity: Combinatorial pharmacogenomics predicts antidepressant responses and healthcare utilizations better than single gene phenotypes. Pharmacogenomics J. 2015;15:443–51. doi: 10.1038/tpj.2014.85.
    1. Winner JG, Carhart JM, Altar CA, Allen JD, Dechairo BM. A prospective, randomized, double-blind study assessing the clinical impact of integrated pharmacogenomic testing for major depressive disorder. Disco Med. 2013;16:219–27.
    1. Hall-Flavin DK, Winner JG, Allen JD, Carhart JM, Proctor B, Snyder KA, et al. Utility of integrated pharmacogenomic testing to support the treatment of major depressive disorder in a psychiatric outpatient setting. Pharmacogenetics Genomics. 2013;23:535–48. doi: 10.1097/FPC.0b013e3283649b9a.
    1. Greden JF, Parikh SV, Rothschild AJ, Thase ME, Dunlop BW, DeBattista C, et al. Impact of pharmacogenomics on clinical outcomes in major depressive disorder in the GUIDED trial: A large, patient- and rater-blinded, randomized, controlled study. J Psychiatr Res. 2019;111:59–67. doi: 10.1016/j.jpsychires.2019.01.003.
    1. Tanner JA, Davies PE, Voudouris NC, Shahmirian A, Herbert D, Braganza N, et al. Combinatorial pharmacogenomics and improved patient outcomes in depression: Treatment by primary care physicians or psychiatrists. J Psychiatr Res. 2018;104:157–62. doi: 10.1016/j.jpsychires.2018.07.012.
    1. Lasser KE, Himmelstein DU, Woolhandler S. Access to care, health status, and health disparities in the United States and Canada: results of a cross-national population-based survey. Am J Public Health. 2006;96:1300–7. doi: 10.2105/AJPH.2004.059402.
    1. Tanner JA, Brown LC, Yu K, Li J, Dechairo BM. Canadian medication cost savings associated with combinatorial pharmacogenomic guidance for psychiatric medications. Clinicoecon Outcomes Res. 2019;11:779–87. doi: 10.2147/CEOR.S224277.
    1. Tanner JA, Davies PE, Overall CC, Grima D, Nam J, Dechairo BM. Cost-effectiveness of combinatorial pharmacogenomic testing for depression from the Canadian public payer perspective. Pharmacogenomics. 2020;21:521–31. doi: 10.2217/pgs-2020-0012.
    1. Jablonski M, King N, Wang Y, Winner JG, Watterson LR, Gunselman S, et al. Analytical validation of a psychiatric pharmacogenomic test. Personalized Medicine. 2018;15:189–197.
    1. Malhotra AK, Correll CU, Chowdhury NI, Müller DJ, Gregersen PK, Lee AT, et al. Association between common variants near the melanocortin 4 receptor gene and severe antipsychotic drug–induced weight gain. Arch Gen Psychiatry. 2012;69:904–12. doi: 10.1001/archgenpsychiatry.2012.191.
    1. Tiwari A, Zai C, Likhodi O, Lisker A, Singh D, Souza R, et al. Association of a common polymorphism in the Cannabinoid receptor 1 (CNR1) gene with antipsychotic-induced weight gain in chronic schizophrenia subjects. Neuropsychopharmacol: Off Publ Am Coll Neuropsychopharmacol. 2010;35:1315–24. doi: 10.1038/npp.2009.235.
    1. Tiwari AK, Brandl EJ, Weber C, Likhodi O, Zai CC, Hahn MK, et al. Association of a functional polymorphism in neuropeptide Y with antipsychotic-induced weight gain in schizophrenia patients. J Clin Psychopharmacol. 2013;33:11–17. doi: 10.1097/JCP.0b013e31827d145a.
    1. Brandl EJ, Tiwari AK, Chowdhury NI, Zai CC, Lieberman JA, Meltzer HY, et al. Genetic variation in the GCG and in the GLP1R genes and antipsychotic-induced weight gain. Pharmacogenomics. 2014;15:423–31. doi: 10.2217/pgs.13.247.
    1. Tiwari AK, Brandl EJ, Zai CC, Goncalves VF, Chowdhury NI, Freeman N, et al. Association of orexin receptor polymorphisms with antipsychotic-induced weight gain. World J Biol Psychiatry. 2016;17:221–9. doi: 10.3109/15622975.2015.1076173.
    1. Gonçalves VF, Zai CC, Tiwari AK, Brandl EJ, Derkach A, Meltzer HY, et al. A hypothesis-driven association study of 28 nuclear-encoded mitochondrial genes with antipsychotic-induced weight gain in schizophrenia. Neuropsychopharmacol: Off Publ Am Coll Neuropsychopharmacol. 2014;39:1347–54. doi: 10.1038/npp.2013.312.
    1. Kennedy SH, Lam RW, McIntyre RS, Tourjman SV, Bhat V, Blier P, et al. Canadian network for mood and anxiety treatments (CANMAT) 2016 clinical guidelines for the management of adults with major depressive disorder: Section 3. pharmacological treatments. Can J Psychiatry Rev Canadienne de Psychiatr. 2016;61:540–60.
    1. American Psychiatric Association. Practice Guidelines. 2020; .
    1. Lam RW, McIntosh D, Wang J, Enns MW, Kolivakis T, Michalak EE, et al. Canadian network for mood and anxiety treatments (CANMAT) 2016 clinical guidelines for the management of adults with major depressive disorder: Section 1. Disease burden and principles of care. Can J Psychiatry Rev Canadienne de Psychiatr. 2016;61:510–23.
    1. Dunlop BW, Parikh SV, Rothschild AJ, Thase ME, DeBattista C, Conway CR, et al. Comparing sensitivity to change using the 6-item versus the 17-item Hamilton depression rating scale in the GUIDED randomized controlled trial. BMC Psychiatry. 2019;19:420. doi: 10.1186/s12888-019-2410-2.
    1. Thase ME, Parikh SV, Rothschild AJ, Dunlop BW, DeBattista C, Conway CR, et al. Impact of pharmacogenomics on clinical outcomes for patients taking medications with gene-drug interactions in a randomized, controlled trial. J Clin Psychiatry. 2019;80:19m12910.
    1. U.S. Department of Commerce. United States Census Beurau: Quick Facts 2019; .
    1. Statistics Canada. Data Tables, 2016 Census 2019; .

Source: PubMed

3
購読する