A Pharmacogenomic-based Antidepressant Treatment for Patients with Major Depressive Disorder: Results from an 8-week, Randomized, Single-blinded Clinical Trial

Changsu Han, Sheng-Min Wang, Won-Myong Bahk, Soo-Jung Lee, Ashwin A Patkar, Prakash S Masand, Laura Mandelli, Chi-Un Pae, Alessandro Serretti, Changsu Han, Sheng-Min Wang, Won-Myong Bahk, Soo-Jung Lee, Ashwin A Patkar, Prakash S Masand, Laura Mandelli, Chi-Un Pae, Alessandro Serretti

Abstract

Objective: Pharmacogenomic-based antidepressant treatment (PGATx) may result in more precise pharmacotherapy of major depressive disorder (MDD) with better drug therapy guidance.

Methods: An 8-week, randomized, single-blind clinical trial was conducted to evaluate the effectiveness and tolerability of PGATx in 100 patients with MDD. All recruited patients were randomly allocated either to PGATx (n=52) or treatment as usual (TAU, n=48) groups. The primary endpoint was a change of total score of the Hamilton Depression Rating Scale-17 (HAMD-17) from baseline to end of treatment. Response rate (at least 50% reduction in HAMD-17 score from baseline), remission rate (HAMD-17 score ≤7 at the end of treatment) as well as the change of total score of Frequency, Intensity, and Burden of Side Effects Ratings (FIBSER) from baseline to end of treatment were also investigated.

Results: The mean change of HAMD-17 score was significantly different between two groups favoring PGATx by -4.1 point of difference (p=0.010) at the end of treatment. The mean change in the FIBSER score from baseline was significantly different between two treatment groups favoring PGATx by -2.5 point of difference (p=0.028). The response rate (71.7 % vs. 43.6%, p=0.014) were also significantly higher in PGATx than in TAU at the end of treatment, while the remission rate was numerically higher in PGATx than in TAU groups without statistical difference (45.5% vs. 25.6%, p=0.071). The reason for early drop-out associated with adverse events was also numerically higher in TAU (n=9, 50.0%) than in PGATx (n=4, 30.8%).

Conclusion: The present study clearly demonstrate that PGATx may be a better treatment option in the treatment of MDD in terms of effectiveness and tolerability; however, study shortcomings may limit a generalization. Adequately-powered, well-designed, subsequent studies should be mandatory to prove its practicability and clinical utility for routine practice.

Keywords: Antidepressants; Depressive disorder; Effects; Pharmacogenetic testing; Precision medicine; Tolerance.

Figures

Fig. 1
Fig. 1
The disposition of the subjects during the study.
Fig. 2
Fig. 2
The response and remission rates between the two treatment groups at the end of treatment. PGATx, pharmacogenomic-based antidepressant treatment; TAU, treatment as usual.
Fig. 3
Fig. 3
The proportion of patients those achieved 2 or less in Frequency, Intensity, and Burden of Side Effects Ratings sub-scores at the end of treatment. PGATx, pharmacogenomic-based antidepressant treatment; TAU, treatment as usual.

References

    1. Rakesh G, Pae CU, Masand PS. Beyond serotonin: newer anti-depressants in the future. Expert Rev Neurother. 2017;17:777–790. doi: 10.1080/14737175.2017.1341310.
    1. Wang SM, Han C, Pae CU. Criticisms of drugs in early development for the treatment of depression: what can be improved? Expert Opin Investig Drugs. 2015;24:445–453. doi: 10.1517/13543784.2014.985784.
    1. Marks DM, Pae CU, Patkar AA. Triple reuptake inhibitors: a premise and promise. Psychiatry Investig. 2008;5:142–147. doi: 10.4306/pi.2008.5.3.142.
    1. Wang HR, Bahk WM, Seo JS, Woo YS, Park YM, Jeong JH, et al. Korean Medication Algorithm for Depressive Disorder: comparisons with other treatment guidelines. Clin Psychopharmacol Neurosci. 2017;15:199–209. doi: 10.9758/cpn.2017.15.3.199.
    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;163:1905–1917. doi: 10.1176/ajp.2006.163.11.1905.
    1. Rush AJ, Warden D, Wisniewski SR, Fava M, Trivedi MH, Gaynes BN, et al. STAR*D: revising conventional wisdom. CNS Drugs. 2009;23:627–647.
    1. Gaynes BN, Warden D, Trivedi MH, Wisniewski SR, Fava M, Rush AJ. What did STAR*D teach us? Results from a large-scale, practical, clinical trial for patients with depression. Psychiatr Serv. 2009;60:1439–1445. doi: 10.1176/ps.2009.60.11.1439.
    1. Pae CU, Wang SM, Han C, Lee SJ, Patkar AA, Masand PS, et al. Vortioxetine: a meta-analysis of 12 short-term, randomised, placebo-controlled clinical trials for the treatment of major depressive disorder. J Psychiatry Neurosci. 2015;40:174–186. doi: 10.1503/jpn.140120.
    1. Cipriani A, Furukawa TA, Salanti G, Chaimani A, Atkinson LZ, Ogawa Y, et al. Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: a systematic review and network meta-analysis. Lancet. 2018;391:1357–1366. doi: 10.1016/S0140-6736(17)32802-7.
    1. Hung CI. Factors predicting adherence to antidepressant treatment. Curr Opin Psychiatry. 2014;27:344–349. doi: 10.1097/YCO.0000000000000086.
    1. Tansey KE, Guipponi M, Hu X, Domenici E, Lewis G, Malafosse A, et al. Contribution of common genetic variants to antidepressant response. Biol Psychiatry. 2013;73:679–682. doi: 10.1016/j.biopsych.2012.10.030.
    1. Licinio J, Wong ML. Pharmacogenomics of antidepressant treatment effects. Dialogues Clin Neurosci. 2011;13:63–71.
    1. Herbert D, Neves-Pereira M, Baidya R, Cheema S, Groleau S, Shahmirian A, et al. Genetic testing as a supporting tool in prescribing psychiatric medication: design and protocol of the IMPACT study. J Psychiatr Res. 2018;96:265–272. doi: 10.1016/j.jpsychires.2017.09.002.
    1. Niitsu T, Fabbri C, Bentini F, Serretti A. Pharmacogenetics in major depression: a comprehensive meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2013;45:183–194. doi: 10.1016/j.pnpbp.2013.05.011.
    1. Drozda K, Müller DJ, Bishop JR. Pharmacogenomic testing for neuropsychiatric drugs: current status of drug labeling, guidelines for using genetic information, and test options. Pharmacotherapy. 2014;34:166–184. doi: 10.1002/phar.1398.
    1. Schuck RN, Grillo JA. Pharmacogenomic biomarkers: an FDA perspective on utilization in biological product labeling. AAPS J. 2016;18:573–577. doi: 10.1208/s12248-016-9891-4.
    1. Frueh FW, Amur S, Mummaneni P, Epstein RS, Aubert RE, DeLuca TM, et al. Pharmacogenomic biomarker information in drug labels approved by the United States food and drug administration: prevalence of related drug use. Pharmacotherapy. 2008;28:992–998. doi: 10.1592/phco.28.8.992.
    1. Winner JG, Carhart JM, Altar CA, Goldfarb S, Allen JD, Lavezzari G, et al. Combinatorial pharmacogenomic guidance for psychiatric medications reduces overall pharmacy costs in a 1 year prospective evaluation. Curr Med Res Opin. 2015;31:1633–1643. doi: 10.1185/03007995.2015.1063483.
    1. Nassan M, Nicholson WT, Elliott MA, Rohrer Vitek CR, Black JL, Frye MA. Pharmacokinetic pharmacogenetic prescribing guidelines for antidepressants: a template for psychiatric precision medicine. Mayo Clin Proc. 2016;91:897–907. doi: 10.1016/j.mayocp.2016.02.023.
    1. de Leon J, Armstrong SC, Cozza KL. Clinical guidelines for psychiatrists for the use of pharmacogenetic testing for CYP450 2D6 and CYP450 2C19. Psychosomatics. 2006;47:75–85. doi: 10.1176/appi.psy.47.1.75.
    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. Hafner S, Haubensak S, Paul T, Zolk O. [How to individualize drug therapy based on pharmacogenetic information? A systematic review of published guidelines]. Dtsch Med Wochenschr. 2016;141:e183–e202. doi: 10.1055/s-0042-100973. German.
    1. Jürgens G, Jacobsen CB, Rasmussen HB, Werge T, Nordentoft M, Andersen SE. Utility and adoption of CYP2D6 and CYP2C19 genotyping and its translation into psychiatric clinical practice. Acta Psychiatr Scand. 2012;125:228–237. doi: 10.1111/j.1600-0447.2011.01802.x.
    1. Benitez J, Jablonski MR, Allen JD, Winner JG. The clinical validity and utility of combinatorial pharmacogenomics: enhancing patient outcomes. Appl Transl Genom. 2015;5:47–49. doi: 10.1016/j.atg.2015.03.001.
    1. Fabbri C, Serretti A. Clinical application of antidepressant pharmacogenetics: considerations for the design of future studies. Neurosci Lett. 2018 doi: 10.1016/j.neulet.2018.06.020. doi: 10.1016/j.neulet.2018.06.020. [Epub ahead of print]
    1. Bousman CA, Dunlop BW. Genotype, phenotype, and medication recommendation agreement among commercial pharmacogenetic-based decision support tools. Pharmacogenomics J. 2018;18:613–622. doi: 10.1038/s41397-018-0027-3.
    1. Bousman CA, Forbes M, Jayaram M, Eyre H, Reynolds CF, Berk M, et al. Antidepressant prescribing in the precision medicine era: a prescriber’s primer on pharmacogenetic tools. BMC Psychiatry. 2017;17:60. doi: 10.1186/s12888-017-1230-5.
    1. Bousman CA, Hopwood M. Commercial pharmacogenetic-based decision-support tools in psychiatry. Lancet Psychiatry. 2016;3:585–590. doi: 10.1016/S2215-0366(16)00017-1.
    1. Zeier Z, Carpenter LL, Kalin NH, Rodriguez CI, McDonald WM, Widge AS, et al. Clinical implementation of pharmacogenetic decision support tools for antidepressant drug prescribing. Am J Psychiatry. 2018;175:873–886. doi: 10.1176/appi.ajp.2018.17111282.
    1. Bousman CA, Müller DJ, Ng CH, Byron K, Berk M, Singh AB. Concordance between actual and pharmacogenetic predicted desvenlafaxine dose needed to achieve remission in major depressive disorder: a 10-week open-label study. Pharmacogenet Genomics. 2017;27:1–6. doi: 10.1097/FPC.0000000000000253.
    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;15:443–451. doi: 10.1038/tpj.2014.85.
    1. Pérez V, Salavert A, Espadaler J, Tuson M, Saiz-Ruiz J, Sáez-Navarro C, et al. Efficacy of prospective pharmacogenetic testing in the treatment of major depressive disorder: results of a randomized, double-blind clinical trial. BMCPsychiatry. 2017;17:250.
    1. Espadaler J, Tuson M, Lopez-Ibor JM, Lopez-Ibor F, Lopez-Ibor MI. Pharmacogenetic testing for the guidance of psychiatric treatment: a multicenter retrospective analysis. CNS Spectr. 2017;22:315–324. doi: 10.1017/S1092852915000711.
    1. Bradley P, Shiekh M, Mehra V, Vrbicky K, Layle S, Olson MC, et al. Improved efficacy with targeted pharmacogenetic-guided treatment of patients with depression and anxiety: a randomized clinical trial demonstrating clinical utility. J Psychiatr Res. 2018;96:100–107. doi: 10.1016/j.jpsychires.2017.09.024.
    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. Discov Med. 2013;16:219–227.
    1. Singh AB. Improved antidepressant remission in major depression via a pharmacokinetic pathway polygene pharmaco-genetic report. Clin Psychopharmacol Neurosci. 2015;13:150–156. doi: 10.9758/cpn.2015.13.2.150.
    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. Pharmacogenet Genomics. 2013;23:535–548. doi: 10.1097/FPC.0b013e3283649b9a.
    1. Brennan FX, Gardner KR, Lombard J, Perlis RH, Fava M, Harris HW, et al. A naturalistic study of the effectiveness of pharmacogenetic testing to guide treatment in psychiatric patients with mood and anxiety disorders. Prim Care Companion CNS Disord. 2015;17
    1. Kennedy SH, Lam RW, Cohen NL, Ravindran AV Group CDW. Clinical guidelines for the treatment of depressive disorders. IV. Medications and other biological treatments. Can J Psychiatry. 2001;46(Suppl 1):38S–58S.
    1. Anderson IM, Ferrier IN, Baldwin RC, Cowen PJ, Howard L, Lewis G, et al. Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 2000 British association for psychopharmacology guidelines. J Psychopharmacol. 2008;22:343–396. doi: 10.1177/0269881107088441.
    1. Bauer M, Pfennig A, Severus E, Whybrow PC, Angst J, Möller HJ. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for biological treatment of unipolar depressive disorders, part 1: update 2013 on the acute and continuation treatment of unipolar depressive disorders. World J Biol Psychiatry. 2013;14:334–385. doi: 10.3109/15622975.2013.804195.
    1. Patkar AA, Pae CU. Atypical antipsychotic augmentation strategies in the context of guideline-based care for the treatment of major depressive disorder. CNS Drugs. 2013;27(Suppl 1):S29–S37. doi: 10.1007/s40263-012-0031-0.
    1. Rush AJ, Trivedi MH, Stewart JW, Nierenberg AA, Fava M, Kurian BT, et al. Combining medications to enhance depression outcomes (CO-MED): acute and long-term outcomes of a single-blind randomized study. Am J Psychiatry. 2011;168:689–701. doi: 10.1176/appi.ajp.2011.10111645.
    1. Pae CU, Patkar AA. Clinical issues in use of atypical anti-psychotics for depressed patients. CNS Drugs. 2013;27(Suppl 1):S39–S45. doi: 10.1007/s40263-012-0032-z.
    1. Han C, Yeh TL, Kato M, Sato S, Chang CM, Pae CU. Management of chronic depressive patients with residual symptoms. CNS Drugs. 2013;27(Suppl 1):S53–S57. doi: 10.1007/s40263-012-0034-x.
    1. Han C, Wang SM, Kato M, Lee SJ, Patkar AA, Masand PS, et al. Second-generation antipsychotics in the treatment of major depressive disorder: current evidence. Expert Rev Neurother. 2013;13:851–870. doi: 10.1586/14737175.2013.811901.
    1. Torrellas C, Carril JC, Cacabelos R. Optimization of anti-depressant use with pharmacogenetic strategies. Curr Genomics. 2017;18:442–449. doi: 10.2174/1389202918666170426164940.
    1. Jeon SW, Han C, Ko YH, Yoon SY, Pae CU, Choi J, et al. Measurement-based treatment of residual symptoms using clinically useful depression outcome scale: Korean validation study. Clin Psychopharmacol Neurosci. 2017;15:28–34. doi: 10.9758/cpn.2017.15.1.28.
    1. Serretti A, Kato M, De Ronchi D, Kinoshita T. Meta-analysis of serotonin transporter gene promoter polymorphism (5-HTTLPR) association with selective serotonin reuptake inhibitor efficacy in depressed patients. Mol Psychiatry. 2007;12:247–257. doi: 10.1038/sj.mp.4001926.
    1. Reyes-Barron C, Tonarelli S, Delozier A, Briones DF, Su BB, Rubin LP, et al. Pharmacogenetics of antidepressants, a review of significant genetic variants in different populations. Clin Depress. 2016;2:1–10.
    1. Hornberger J, Li Q, Quinn B. Cost-effectiveness of combinatorial pharmacogenomic testing for treatment-resistant major depressive disorder patients. Am J Manag Care. 2015;21:e357–e365.
    1. Najafzadeh M, Garces JA, Maciel A. Economic evaluation of implementing a novel pharmacogenomic test (IDgenetix®) to guide treatment of patients with depression and/or anxiety. Pharmacoeconomics. 2017;35:1297–1310. doi: 10.1007/s40273-017-0587-0.
    1. Verbelen M, Weale ME, Lewis CM. Cost-effectiveness of pharmacogenetic–guided treatment: are we there yet? Pharmacogenomics J. 2017;17:395–402. doi: 10.1038/tpj.2017.21.
    1. Stahl SM. Psychiatric pharmacogenomics: how to integrate into clinical practice. CNS Spectr. 2017;22:1–4. doi: 10.1017/S109285291600095X.
    1. Perlis RH. Cytochrome P450 genotyping and antidepressants. BMJ. 2007;334:759. doi: 10.1136/bmj.39169.547512.80.

Source: PubMed

3
구독하다