Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-c47g7 Total loading time: 0 Render date: 2024-04-19T16:54:54.661Z Has data issue: false hasContentIssue false

Part 2 - Psychopharmacology of the Main Psychotropic Drug Groups

Published online by Cambridge University Press:  29 May 2020

Peter M. Haddad
Affiliation:
Hamad Medical Corporation, Qatar
David J. Nutt
Affiliation:
Centre for Neuropsychopharmacology, Division of Psychiatry, Department of Brain Sciences, Imperial College London
Get access
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2020

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Aan Het Rot, M, Zarate, CA Jr, Charney, DS, Mathew, SJ (2012). Ketamine for depression: where do we go from here? Biol Psychiatry, 72(7), 537547. doi:10.1016/j.biopsych.2012.05.003.CrossRefGoogle Scholar
Aberg-Wistedt, A, Agren, H, Ekselius, L, Bengtsson, F, Akerblad, AC (2000). Sertraline versus paroxetine in major depression: clinical outcome after six months of continuous therapy. J Clin Psychopharmacol, 20, 645652.Google Scholar
Adli, M, Baethge, C, Heinz, A, Langlitz, N, Bauer, M (2005). Is dose escalation of antidepressants a rational strategy after a medium-dose treatment has failed? A systematic review. Eur Arch Psychiatry Clin Neurosci, 255, 387400.CrossRefGoogle ScholarPubMed
Akerblad, AC, Bengtsson, F, von Knorring, L, Ekselius, L (2006). Response, remission and relapse in relation to adherence in primary care treatment of depression: a 2-year outcome study. Int Clin Psychopharmacol, 21, 117124.Google Scholar
Alvarez, E, Perez, V, Artigas, F (2014). Pharmacology and clinical potential of vortioxetine in the treatment of major depressive disorder. Neuropsychiatr Dis Treat, 10, 12971307.Google Scholar
American Psychiatric Association (2013). Diagnostic and Statistical Manual of Mental Disorders, 5th ed. Arlington, VA: American Psychiatric Association.Google Scholar
Anderson, IM (2000). Selective serotonin reuptake inhibitors versus tricyclic antidepressants: a meta-analysis of efficacy and tolerability. J Affect Disord, 58, 1936.CrossRefGoogle ScholarPubMed
Anderson, IM (2001). Meta-analytical studies on new antidepressants. Br Med Bull, 57, 161178.Google Scholar
Anderson, IM (2003). Drug treatment of depression: reflections on the evidence. Adv Psychiatr Treat, 9, 1120.CrossRefGoogle Scholar
Anglin, R, Yuan, Y, Moayyedi, P, et al. (2014). Risk of upper gastrointestinal bleeding with selective serotonin reuptake inhibitors with or without concurrent nonsteroidal anti-inflammatory use: a systematic review and meta-analysis. Am J Gastroenterol, 109(6), 811819. doi:10.1038/ajg.2014.82.CrossRefGoogle ScholarPubMed
Angst, J, Scheidegger, P, Stabl, M (1993). Efficacy of moclobemide in different patient groups. Results of new subscales of the Hamilton Depression Rating Scale. Clin Neuropharmacol, 16(Suppl 2), S55S62.Google ScholarPubMed
Angst, J, Gamma, A, Bowden, CL, et al. (2012). Diagnostic criteria for bipolarity based on an international sample of 5,635 patients with DSM-IV major depressive episodes. Eur Arch Psychiatry Clin Neurosci, 262(1), 311. doi:10.1007/s00406-011-0228-0.CrossRefGoogle Scholar
Appelberg, BG, Syvalahti, EK, Koskinen, TE, et al. (2001). Patients with severe depression may benefit from buspirone augmentation of selective serotonin reuptake inhibitors: results from a placebo-controlled, randomized, double-blind, placebo wash-in study. J Clin Psychiatry, 62, 448452.CrossRefGoogle ScholarPubMed
Aronson, R, Offman, HJ, Joffe, RT, Naylor, CD (1996). Triiodothyronine augmentation in the treatment of refractory depression. A meta-analysis. Arch Gen Psychiatry, 53, 842848.Google Scholar
Bair, MJ, Robinson, RL, Eckert, GJ, et al. (2004). Impact of pain on depression treatment response in primary care. Psychosom Med, 66, 1722.CrossRefGoogle ScholarPubMed
Baldessarini, RJ, Tondo, L, Ghiana, C, Lepri, B (2010). Illness risk following rapid versus gradual discontinuation of antidepressants. Am J Psychiatry, 167, 934994.Google Scholar
Baldwin, DS, Anderson, IM, Nutt, DJ, et al. (2005). Evidence-based guidelines for the pharmacological treatment of anxiety disorders: recommendations from the British Association for Psychopharmacology. J Psychopharmacol, 19, 567596.Google Scholar
Barbee, JG, Thompson, TR, Jamhour, NJ, et al. (2011). A double-blind placebo-controlled trial of lamotrigine as an antidepressant augmentation agent in treatment-refractory unipolar depression. J Clin Psychiatry, 72(10), 14051412. doi:10.4088/JCP.09m05355gre.Google Scholar
Barbosa, L, Berk, M, Vorster, M (2003). A double-blind, randomized, placebo-controlled trial of augmentation with lamotrigine or placebo in patients concomitantly treated with fluoxetine for resistant major depressive episodes. J Clin Psychiatry, 64, 403407.Google Scholar
Bateman, DN (2003). Poisonous substances: antidepressants. Medicine, 31, 3234.CrossRefGoogle Scholar
Bauer, M, Dell’osso, L, Kasper, S, et al. (2013). Extended-release quetiapine fumarate (quetiapine XR) monotherapy and quetiapine XR or lithium as add-on to antidepressants in patients with treatment-resistant major depressive disorder. J Affect Disord, 151(1), 209219. doi:10.1016/j.jad.2013.05.079.Google Scholar
Bauer, M, Adli, M, Ricken, R, Severus, E, Pilhatsch, M (2014). Role of lithium augmentation in the management of major depressive disorder. CNS Drugs, 28(4), 331342. doi:10.1007/s40263-014-0152-8.CrossRefGoogle ScholarPubMed
Baumann, P, Ulrich, S, Eckermann, G, et al. (2005). The AGNP-TDM Expert Group Consensus Guidelines: focus on therapeutic monitoring of antidepressants. Dialogues Clin Neurosci, 7, 231247.Google Scholar
Berman, RM, Marcus, RN, Swanink, R, et al. (2007). The efficacy and safety of aripiprazole as adjunctive therapy in major depressive disorder: a multicenter, randomized, double-blind, placebo-controlled study. J Clin Psychiatry, 68, 843853.Google Scholar
Birkenhager, TK, van den Broek, WW, Moleman, P, Bruijn, JA (2006). Outcome of a 4-step treatment algorithm for depressed inpatients. J Clin Psychiatry, 67, 12661271.CrossRefGoogle ScholarPubMed
Blier, P, Gobbi, G, Turcotte, JE, et al. (2009). Mirtazapine and paroxetine in major depression: a comparison of monotherapy versus their combination from treatment initiation. Eur Neuropsychopharmacol, 19(7), 457465.CrossRefGoogle ScholarPubMed
Blier, P, Ward, HE, Tremblay, P, et al. (2010). Combination of antidepressant medications from treatment initiation for major depressive disorder: a double-blind randomized study. Am J Psychiatry, 167(3), 281288.CrossRefGoogle ScholarPubMed
Bridge, JA, Iyengar, S, Salary, CB, et al. (2007). Clinical response and risk for reported suicidal ideation and suicide attempts in pediatric antidepressant treatment: a meta-analysis of randomized controlled trials. JAMA, 297, 16831696.Google Scholar
Brown, WA (2007). Treatment response in melancholia. Acta Psychiatr Scand, 115(Suppl 433), 125129.Google Scholar
Bryant, SG, Fisher, S, Kluge, RM (1987). Long-term versus short-term amitriptyline side effects as measured by a postmarketing surveillance system. J Clin Psychopharmacol, 7, 7882.Google Scholar
Buckley, NA, McManus, PR (2002). Fatal toxicity of serotoninergic and other antidepressant drugs: analysis of United Kingdom mortality data. BMJ, 325, 13321333.Google Scholar
Bukh, JD, Bock, C, Vinberg, M, Kessing, LV (2013). The effect of prolonged duration of untreated depression on antidepressant treatment outcome. J Affect Disord, 145(1), 4248. doi:10.1016/j.jad.2012.07.008.Google Scholar
Bump, GM, Mulsant, BH, Pollock, BG, et al. (2001). Paroxetine versus nortriptyline in the continuation and maintenance treatment of depression in the elderly. Depress Anxiety, 13, 3844.Google Scholar
Burke, WJ, Gergel, I, Bose, A (2002). Fixed-dose trial of the single isomer SSRI escitalopram in depressed outpatients. J Clin Psychiatry, 63, 331336.Google Scholar
Byrne, SE, Rothschild, AJ (1998). Loss of antidepressant efficacy during maintenance therapy: possible mechanisms and treatments. J Clin Psychiatry, 59, 279288.Google Scholar
Canuso, CM, Singh, JB, Fedgchin, M, et al. (2018). Efficacy and safety of intranasal esketamine for the rapid reduction of symptoms of depression and suicidality in patients at imminent risk for suicide: results of a double-blind, randomized, placebo-controlled study. Am J Psychiatry, 175, 620630. doi:10.1176/appi.ajp.2018.17060720.Google Scholar
Carpenter, LL, Yasmin, S, Price, LH (2002). A double-blind, placebo-controlled study of antidepressant augmentation with mirtazapine. Biol Psychiatry, 51, 183188.Google Scholar
Castro, VM, Clements, CC, Murphy, SN, et al. (2013). QT interval and antidepressant use: a cross sectional study of electronic health records. BMJ, 346, f288. doi:10.1136/bmj.f288.Google Scholar
Cheeta, S, Schifano, F, Oyefeso, A, Webb, L, Ghodse, AH (2004). Antidepressant-related deaths and antidepressant prescriptions in England and Wales, 1998–2000. Br J Psychiatry, 184, 4147.CrossRefGoogle ScholarPubMed
Cheng, YL, Hu, HY, Lin, XH, et al. (2015). Use of SSRI, but not SNRI, increased upper and lower gastrointestinal bleeding: a nationwide population-based cohort study in Taiwan. Medicine (Baltimore), 94(46), e2022. doi:10.1097/MD.0000000000002022.Google Scholar
Cipriani, A, Hawton, K, Stockton, S, Geddes, JR (2013). Lithium in the prevention of suicide in mood disorders: updated systematic review and meta-analysis. BMJ, 346, f3646. https://doi.org/10.1136/bmj.f3646.CrossRefGoogle ScholarPubMed
Cipriani, A, Furukawa, TA, Salanti, G, et al. (2018). 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, 391(10128), 13571366.CrossRefGoogle ScholarPubMed
Cleare, A, Pariante, CM, Young, AH, et al. (2015). Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 2008 British Association for Psychopharmacology guidelines. J Psychopharmacology, 29(5), 459525.Google Scholar
Cramer, JA, Rosenheck, R (1998). Compliance with medication regimens for mental and physical disorders. Psychiatr Serv, 49, 196201.Google Scholar
Crossley, NA, Bauer, M (2007). Acceleration and augmentation of antidepressants with lithium for depressive disorders: two meta-analyses of randomized, placebo-controlled trials. J Clin Psychiatry, 68, 935940.Google Scholar
Dawson, R, Lavori, PW, Coryell, WH, Endicott, J, Keller, MB (1998). Maintenance strategies for unipolar depression: an observational study of levels of treatment and recurrence. J Affect Disord, 49, 3144.Google Scholar
de Abajo, FJ, Garcia-Rodriguez, LA (2008). Risk of upper gastrointestinal tract bleeding associated with selective serotonin reuptake inhibitors and venlafaxine therapy: interaction with nonsteroidal anti-inflammatory drugs and effect of acid-suppressing agents. Arch Gen Psychiatry, 65, 795803.Google Scholar
Delgado, PL (2004). Common pathways of depression and pain. J Clin Psychiatry, 65(Suppl 12), 1619.Google Scholar
Delgado, PL, Brannan, SK, Mallinckrodt, CH, et al. (2005). Sexual functioning assessed in 4 double-blind placebo- and paroxetine-controlled trials of duloxetine for major depressive disorder. J Clin Psychiatry, 66, 686692.Google Scholar
Demyttenaere, K, Albert, A, Mesters, P, et al. (2005). What happens with adverse events during 6 months of treatment with selective serotonin reuptake inhibitors? J Clin Psychiatry, 66, 859863.Google Scholar
Detke, MJ, Wiltse, CG, Mallinckrodt, CH, et al. (2004). Duloxetine in the acute and long-term treatment of major depressive disorder: a placebo- and paroxetine-controlled trial. Eur Neuropsychopharmacol, 14, 457470.CrossRefGoogle ScholarPubMed
DeVeaugh-Geiss, AM, West, SL, Miller, WC, et al. (2010). The adverse effects of comorbid pain on depression outcomes in primary care patients: results from the ARTIST trial. Pain Med, 11(5), 732741. doi:10.1111/j.1526-4637.2010.00830.x.CrossRefGoogle ScholarPubMed
Dombrovski, AY, Mulsant, BH, Houck, PR, et al. (2007). Residual symptoms and recurrence during maintenance treatment of late-life depression. J Affect Disord, 103, 7782.CrossRefGoogle ScholarPubMed
Dornseif, BE, Dunlop, SR, Potvin, JH, Wernicke, JF (1989). Effect of dose escalation after low-dose fluoxetine therapy. Psychopharmacol Bull, 25, 7179.Google Scholar
Dotoli, D, Spagnolo, C, Bongiorno, F, et al. (2006). Relapse during a 6-month continuation treatment with fluvoxamine in an Italian population: the role of clinical, psychosocial and genetic variables. Prog Neuropsychopharmacol Biol Psychiatry, 30, 442448.CrossRefGoogle Scholar
Dubini, A, Bosc, M, Polin, V (1997). Noradrenaline-selective versus serotonin-selective antidepressant therapy: differential effects on social functioning. J Psychopharmacol, 11, S17S23.Google ScholarPubMed
Edwards, JG, Anderson, I (1999). Systematic review and guide to selection of selective serotonin reuptake inhibitors. Drugs, 57(4), 507533.Google Scholar
El-Khalili, N, Joyce, M, Atkinson, S, et al. (2010). Extended-release quetiapine fumerate (quetiapine XR) as adjunctive therapy in major depressive disorder (MDD) in patients with an inadequate response to ongoing antidepressant treatment: a multicentre, randomized, double-blind, placebo-controlled study. Int J Neuropsychopharmacol, 13, 917932.Google Scholar
Ezquiaga, E, Garcia, A, Bravo, F, Pallares, T (1998). Factors associated with outcome in major depression: a 6-month prospective study. Soc Psychiatry Psychiatr Epidemiol, 33, 552557.CrossRefGoogle ScholarPubMed
Fava, GA, Ruini, C, Rafanelli, C, Grandi, S (2002). Cognitive behavior approach to loss of clinical effect during long-term antidepressant treatment: a pilot study. Am J Psychiatry, 159, 20942095.CrossRefGoogle ScholarPubMed
Fava, M, Rappe, SM, Pava, JA, et al. (1995). Relapse in patients on long-term fluoxetine treatment: response to increased fluoxetine dose. J Clin Psychiatry, 56, 5255.Google Scholar
Fava, M, Uebelacker, LA, Alpert, JE, et al. (1997). Major depressive subtypes and treatment response. Biol Psychiatry, 42, 568576.Google Scholar
Fava, M, Detke, MJ, Balestrieri, M, et al. (2006). Management of depression relapse: re-initiation of duloxetine treatment or dose increase. J Psychiatr Res, 40, 328336.Google Scholar
Fava, M, Thase, ME, DeBattista, C, et al. (2007). Modafinil augmentation of selective serotonin reuptake inhibitor therapy in MDD partial responders with persistent fatigue and sleepiness. Ann Clin Psychiatry, 19, 153159.CrossRefGoogle ScholarPubMed
Fekadu, A, Wooderson, S, Donaldson, C, et al. (2009). A multidimensional tool to quantify treatment resistance in depression: the Maudsley staging method. J Clin Psychiatry, 70, 177184.Google Scholar
Fekadu, A, Donocik, J, Cleare, AJ (2018). Standardisation framework for the Maudsley staging method for treatment resistance in depression. BMC Psychiatry, 18, 100. Available at: https://bmcpsychiatry.biomedcentral.com/articles/10.1186/s12888-018-1679-x.Google Scholar
Fergusson, D, Doucette, S, Glass, KC, et al. (2005). Association between suicide attempts and selective serotonin reuptake inhibitors: systematic review of randomised controlled trials. BMJ, 330, 396.Google Scholar
Ferreri, M, Lavergne, F, Berlin, I, Payan, C, Puech, AJ (2001). Benefits from mianserin augmentation of fluoxetine in patients with major depression non-responders to fluoxetine alone. Acta Psychiatr Scand, 103, 6672.CrossRefGoogle ScholarPubMed
Flint, AJ, Rifat, SL (1998). Two-year outcome of psychotic depression in late life. Am J Psychiatry, 155, 178183.CrossRefGoogle ScholarPubMed
Fond, G, Loundou, A, Rabu, C, et al. (2014). Ketamine administration in depressive disorders: a systematic review and meta-analysis. Psychopharmacology (Berl), 231(18), 36633676. doi:10.1007/s00213-014-3664-5.Google Scholar
Franchini, L, Gasperini, M, Zanardi, R, Smeraldi, E (2000a). Four-year follow-up study of sertraline and fluvoxamine in long-term treatment of unipolar subjects with high recurrence rate. J Affect Disord, 58, 233236.CrossRefGoogle ScholarPubMed
Franchini, L, Rossini, D, Bongiorno, F, et al. (2000b). Will a second prophylactic treatment with a higher dosage of the same antidepressant either prevent or delay new depressive episodes? Psychiatry Res, 96, 8185.CrossRefGoogle ScholarPubMed
Frank, E, Kupfer, DJ, Perel, JM, et al. (1990). Three-year outcomes for maintenance therapies in recurrent depression. Arch Gen Psychiatry, 47, 10931099.Google Scholar
Frank, E, Kupfer, DJ, Perel, JM, et al. (1993). Comparison of full-dose versus half-dose pharmacotherapy in the maintenance treatment of recurrent depression. J Affect Disord, 27, 139145.Google Scholar
Furukawa, TA, McGuire, H, Barbui, C (2002). Meta-analysis of effects and side effects of low dosage tricyclic antidepressants in depression: systematic review. BMJ, 325, 991.Google Scholar
Geddes, JR, Carney, SM, Davies, C, et al. (2003). Relapse prevention with antidepressant drug treatment in depressive disorders: a systematic review. Lancet, 361, 653661.Google Scholar
Ghaziuddin, M, Ghaziuddin, N, Stein, GS (1990). Life events and the recurrence of depression. Can J Psychiatry, 35, 239242.CrossRefGoogle ScholarPubMed
Gibbons, RD, Brown, CH, Hur, K, et al. (2007). Early evidence on the effects of regulators’ suicidality warnings on SSRI prescriptions and suicide in children and adolescents. Am J Psychiatry, 164, 13561363.Google Scholar
Gijsman, HJ, Geddes, JR, Rendell, JM, et al. (2004). Antidepressants for bipolar depression: a systematic review of randomized, controlled trials. Am J Psychiatry, 161 15371547.CrossRefGoogle ScholarPubMed
Goldstein, DJ, Lu, Y, Detke, MJ, et al. (2004). Duloxetine in the treatment of depression: a double-blind placebo-controlled comparison with paroxetine. J Clin Psychopharmacol, 24, 389399.CrossRefGoogle ScholarPubMed
Goodwin, GM, Haddad, PM, Ferrier, IN, et al. (2016). Evidence-based guidelines for treating bipolar disorder: revised third edition recommendations from the British Association for Psychopharmacology. J Psychopharmacol, 30(6), 495553.Google Scholar
Greden, JF (1993). Antidepressant maintenance medications: when to discontinue and how to stop. J Clin Psychiatry, 54 Suppl, 3945.Google Scholar
Greist, J, McNamara, RK, Mallinckrodt, CH, Rayamajhi, JN, Raskin, J (2004). Incidence and duration of antidepressant-induced nausea: duloxetine compared with paroxetine and fluoxetine. Clin Ther, 26, 14461455.Google Scholar
Gunnell, D, Saperia, J, Ashby, D (2005). Selective serotonin reuptake inhibitors (SSRIs) and suicide in adults: meta-analysis of drug company data from placebo controlled, randomised controlled trials submitted to the MHRA’s safety review. BMJ, 330, 385.Google Scholar
Guo, T, Xiang, YT, Xiao, L, et al. (2015). Measurement-based care versus standard care for major depression: a randomized controlled trial with blind raters. Am J Psychiatry, 172(10), 10041013.CrossRefGoogle ScholarPubMed
Gusmao, R, Quintao, S, McDaid, D, et al. (2013). Antidepressant utilization and suicide in Europe: an ecological multi-national study. PLoS One, 8, e66455.Google Scholar
Haddad, PM, Anderson, IM (2002). Antipsychotic-related QTc prolongation, torsade de pointes and sudden death. Drugs, 62, 16491671.Google Scholar
Halperin, D, Reber, G (2007). Influence of antidepressants on hemostasis. Dialogues Clin Neurosci, 9, 4759.Google Scholar
Hammerlein, A, Derendorf, H, Lowenthal, DT (1998). Pharmacokinetic and pharmacodynamic changes in the elderly. Clinical implications. Clin Pharmacokinet, 35, 4964.Google Scholar
Hansen, R, Gaynes, B, Thieda, P, et al. (2008). Meta-analysis of major depressive disorder relapse and recurrence with second-generation antidepressants. Psychiatr Serv, 59(10), 11211130. doi:10.1176/appi.ps.59.10.1121.Google Scholar
Hawton, K, Bergen, H, Simkin, S, et al. (2010). Toxicity of antidepressants: rates of suicide relative to prescribing and non-fatal overdose. Br J Psychiatry, 196(5), 354358. doi:10.1192/bjp.bp.109.070219.Google Scholar
Healy, D, McMonagle, T (1997). The enhancement of social functioning as a therapeutic principle in the management of depression. J Psychopharmacol, 11, S25S31.Google ScholarPubMed
Henkel, V, Mergl, R, Allgaier, AK, et al. (2006). Treatment of depression with atypical features: a meta-analytic approach. Psychiatry Res, 141, 89101.Google Scholar
Hoffman, L, Enders, J, Luo, J, et al. (2003). Impact of an antidepressant management program on medication adherence. Am J Manag Care, 9, 7080.Google Scholar
Howland, RH (2010). Potential adverse effects of discontinuing psychotropic drugs: part 2: antidepressant drugs. J Psychosoc Nurs Ment Health Serv, 48, 912. doi:10.3928/02793695-20100527-98.Google Scholar
Iosifescu, DV, Nierenberg, AA, Alpert, JE, et al. (2004). Comorbid medical illness and relapse of major depressive disorder in the continuation phase of treatment. Psychosomatics, 45, 419425.Google Scholar
Isbister, GK, Bowe, SJ, Dawson, A, Whyte, IM (2004). Relative toxicity of selective serotonin reuptake inhibitors (SSRIs) in overdose. J Toxicol Clin Toxicol, 42, 277285.Google Scholar
Jacobsen, PL, Mahableshwarkar, AR, Palo, WA, et al. (2016). Treatment-emergent sexual dysfunction in randomized trials of vortioxetine for major depressive disorder or generalized anxiety disorder: a pooled analysis. CNS Spectr, 21, 367378.Google Scholar
Jick, H, Kaye, JA, Jick, SS (2004). Antidepressants and the risk of suicidal behaviors. JAMA, 292, 338343.Google Scholar
Joint Formulary Committee (2018). British National Formulary, 75th ed. London: BMJ Group and Pharmaceutical Press.Google Scholar
Kanai, T, Takeuchi, H, Furukawa, TA, et al. (2003). Time to recurrence after recovery from major depressive episodes and its predictors. Psychol Med, 33, 839845.Google Scholar
Karp, JF, Scott, J, Houck, P, et al. (2005). Pain predicts longer time to remission during treatment of recurrent depression. J Clin Psychiatry, 66, 591597.Google Scholar
Katz, MM, Tekell, JL, Bowden, CL, et al. (2004). Onset and early behavioral effects of pharmacologically different antidepressants and placebo in depression. Neuropsychopharmacology, 29, 566579.Google Scholar
Kessing, LV (1998). Recurrence in affective disorder. II. Effect of age and gender. Br J Psychiatry, 172, 2934.CrossRefGoogle ScholarPubMed
Kessing, LV (2003). Subtypes of depressive episodes according to ICD-10: prediction of risk of relapse and suicide. Psychopathology, 36, 285291.Google Scholar
Kessing, LV (2007). Epidemiology of subtypes of depression. Acta Psychiatr Scand, 433 Suppl, 8589.Google Scholar
Kessing, LV, Andersen, PK (2005). Predictive effects of previous episodes on the risk of recurrence in depressive and bipolar disorders. Curr Psychiatry Rep, 7, 413420.Google Scholar
Kessler, D, Burns, A, Tallon, D, et al. (2018). Combining mirtazapine with SSRIs or SNRIs for treatment-resistant depression: the MIR RCT. Health Technol Assess, 22(63), 1136.CrossRefGoogle ScholarPubMed
Köhler-Forsberg, O, Lydholm, N, Hjorthøj, C, et al. (2019). Efficacy of anti-inflammatory treatment on major depressive disorder or depressive symptoms: meta-analysis of clinical trials. Acta Psychiatr Scand, 139, 404419.CrossRefGoogle Scholar
Lader, M (1983). Combined use of tricyclic antidepressants and monoamine oxidase inhibitors. J Clin Psychiatry, 44, 2024.Google ScholarPubMed
Lam, RW, Gorman, CP, Michalon, M, et al. (1995). Multicenter, placebo-controlled study of fluoxetine in seasonal affective disorder. Am J Psychiatry, 152, 17651770.Google Scholar
Landen, M, Bjorling, G, Agren, H, Fahlen, T (1998). A randomized, double-blind, placebo-controlled trial of buspirone in combination with an SSRI in patients with treatment-refractory depression. J Clin Psychiatry, 59, 664668.CrossRefGoogle ScholarPubMed
Lee, P, Shu, L, Xu, X, et al. (2007). Once-daily duloxetine 60 mg in the treatment of major depressive disorder: multicenter, double-blind, randomized, paroxetine-controlled, non-inferiority trial in China, Korea, Taiwan and Brazil. Psychiatry Clin Neurosci, 61, 295307.Google Scholar
Leinonen, E, Skarstein, J, Behnke, K, Agren, H, Helsdingen, JT (1999). Efficacy and tolerability of mirtazapine versus citalopram: a double-blind, randomized study in patients with major depressive disorder. Nordic Antidepressant Study Group. Int Clin Psychopharmacol, 14, 329337.Google Scholar
Leucht, S, Hierl, S, Kissling, W, Dold, M, Davis, JM (2012). Putting the efficacy of psychiatric and general medicine medication into perspective: review of meta-analyses. Br J Psychiatry, 200(2), 97106. doi:10.1192/bjp.bp.111.096594.Google Scholar
Leuchter, AF, Husain, MM, Cook, IA, et al. (2010). Painful physical symptoms and treatment outcome in major depressive disorder: a STAR*D (Sequenced Treatment Alternatives to Relieve Depression) report. Psychol Med, 40(2), 239251. doi:10.1017/S0033291709006035.Google Scholar
Licht, RW, Qvitzau, S (2002). Treatment strategies in patients with major depression not responding to first-line sertraline treatment. A randomised study of extended duration of treatment, dose increase or mianserin augmentation. Psychopharmacology (Berl), 161, 143151.Google Scholar
Lonnqvist, J, Sihvo, S, Syvalahti, E, et al. (1995). Moclobemide and fluoxetine in the prevention of relapses following acute treatment of depression. Acta Psychiatr Scand, 91, 189194.CrossRefGoogle ScholarPubMed
Maes, M, Libbrecht, I, van Hunsel, F, Campens, D, Meltzer, HY (1999). Pindolol and mianserin augment the antidepressant activity of fluoxetine in hospitalized major depressed patients, including those with treatment resistance. J Clin Psychopharmacol, 19, 177182.Google Scholar
Maidment, R, Livingston, G, Katona, C (2002). Just keep taking the tablets: adherence to antidepressant treatment in older people in primary care. Int J Geriatr Psychiatry, 17(8), 752757.Google Scholar
Masand, PS, Gupta, S (2002). Long-term side effects of newer-generation antidepressants: SSRIs, venlafaxine, nefazodone, bupropion, and mirtazapine. Ann Clin Psychiatry, 14, 175182.Google Scholar
Mazure, CM, Bruce, ML, Maciejewski, PK, Jacobs, SC (2000). Adverse life events and cognitive-personality characteristics in the prediction of major depression and antidepressant response. Am J Psychiatry, 157, 896903.Google Scholar
McAllister-Williams, RH, Christmas, D, Cleare, AJ, et al. (2018). Multiple-therapy-resistant-major depressive disorder: a clinically important concept. Br J Psychiatry, 212(5), 274278. doi:10.1192/bjp.2017.33.Google Scholar
McGirr, A, Berlim, MT, Bond, DJ, et al. (2015). A systematic review and meta-analysis of randomized, double-blind, placebo-controlled trials of ketamine in the rapid treatment of major depressive episodes. Psychol Med, 45(4), 693704. doi:10.1017/S0033291714001603.Google Scholar
McGrath, PJ, Stewart, JW, Fava, M, et al. (2006a). Tranylcypromine versus venlafaxine plus mirtazapine following three failed antidepressant medication trials for depression: a STAR*D report. Am J Psychiatry, 163, 15311541.Google Scholar
McGrath, PJ, Stewart, JW, Quitkin, FM, et al. (2006b). Predictors of relapse in a prospective study of fluoxetine treatment of major depression. Am J Psychiatry, 163, 15421548.Google Scholar
McLaughlin, T, Hogue, SL, Stang, PE (2007). Once-daily bupropion associated with improved patient adherence compared with twice-daily bupropion in treatment of depression. Am J Ther, 14, 221225.Google Scholar
Medhus, A, Heskestad, S, Tjemsland, L (1994). Mianserin added to tricyclic antidepressant in depressed patients not responding to a tricyclic antidepressant alone. A randomised, placebo-controlled, double-blind trial. Nord J Psychiatry, 48, 355358.Google Scholar
Medicines and Healthcare products Regulatory Agency (2011). Citalopram and escitalopram: QT interval prolongation (online). Available at: www.gov.uk/drug-safety-update/citalopram-and-escitalopram-qt-interval-prolongation (last accessed 16.10.19).Google Scholar
Melander, H, Salmonson, T, Abadie, E, et al. (2008). A regulatory Apologia – a review of placebo-controlled studies in regulatory submissions of new-generation antidepressants. Eur Neuropsychopharmacol, 18(9), 623627.Google Scholar
Modell, JG, Rosenthal, NE, Harriett, AE, et al. (2005). Seasonal affective disorder and its prevention by anticipatory treatment with bupropion XL. Biol Psychiatry, 58, 658667.Google Scholar
Montgomery, SA, Reimitz, PE, Zivkov, M (1998). Mirtazapine versus amitriptyline in the long-term treatment of depression: a double-blind placebo-controlled study. Int Clin Psychopharmacol, 13, 6367.CrossRefGoogle ScholarPubMed
Morgan, ML, Cook, IA, Rapkin, AJ, Leuchter, AF (2005). Estrogen augmentation of antidepressants in perimenopausal depression: a pilot study. J Clin Psychiatry, 66, 774780.Google Scholar
Morgan, O, Griffiths, C, Baker, A, Majeed, A (2004). Fatal toxicity of antidepressants in England and Wales, 1993–2002. Health Stat Q, 23, 1824.Google Scholar
Moscovitch, A, Blashko, CA, Eagles, JM, et al. (2004). A placebo-controlled study of sertraline in the treatment of outpatients with seasonal affective disorder. Psychopharmacology (Berl), 171, 390397.Google Scholar
Mueller, TI, Leon, AC, Keller, MB, et al. (1999). Recurrence after recovery from major depressive disorder during 15 years of observational follow-up. Am J Psychiatry, 156, 10001006.Google Scholar
Murrough, JW, Iosifescu, DV, Chang, LC, et al. (2013a). Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am J Psychiatry, 170(10), 11341142. doi:10.1176/appi.ajp.2013.13030392.Google Scholar
Murrough, JW, Perez, AM, Pillemer, S, et al. (2013b). Rapid and longer-term antidepressant effects of repeated ketamine infusions in treatment-resistant major depression. Biol Psychiatry, 74(4), 250256. doi:10.1016/j.biopsych.2012.06.022.Google Scholar
National Institute for Clinical Excellence (2004). Depression: the management of depression in primary and secondary care. Clinical guideline [CG23]. Available at: www.nice.org.uk/guidance/cg23 (last accessed 20.10.19).Google Scholar
National Institute for Health and Clinical Excellence (NICE) (2009). Depression in adults: the treatment and management of depression in adults (update). Clinical guideline [CG90]. London: National Institute for Health and Care Excellence. Available at: www.nice.org.uk/guidance/cg90/chapter/1-guidance (last accessed 16.10.19).Google Scholar
Nelson, JC, Papakostas, GI (2009). Atypical antipsychotic augmentation in major depressive disorder: a meta-analysis of placebo-controlled randomized trials. Am J Psychiatry, 166(9), 980991. doi:10.1176/appi.ajp.2009.09030312.Google Scholar
Nelson, JC, Portera, L, Leon, AC (2005a). Are there differences in the symptoms that respond to a selective serotonin or norepinephrine reuptake inhibitor? Biol Psychiatry, 57, 15351542.Google Scholar
Nelson, JC, Portera, L, Leon, AC (2005b). Residual symptoms in depressed patients after treatment with fluoxetine or reboxetine. J Clin Psychiatry, 66, 14091414.Google Scholar
Nierenberg, AA, McLean, NE, Alpert, JE, et al. (1995). Early nonresponse to fluoxetine as a predictor of poor 8-week outcome. Am J Psychiatry, 152, 15001503.Google ScholarPubMed
Nierenberg, AA, Farabaugh, AH, Alpert, JE, et al. (2000). Timing of onset of antidepressant response with fluoxetine treatment. Am J Psychiatry, 157, 14231428.Google Scholar
Nierenberg, AA, Fava, M, Trivedi, MH, et al. (2006). A comparison of lithium and T(3) augmentation following two failed medication treatments for depression: a STAR*D report. Am J Psychiatry, 163, 15191530.Google Scholar
Nurnberg, H, Hensley, PL, Heiman, JR, et al. (2008). Sildenafil treatment of women with antidepressant-associated sexual dysfunction: a randomized controlled trial. JAMA, 300, 395404.Google Scholar
Ohayon, MM, Schatzberg, AF (2003). Using chronic pain to predict depressive morbidity in the general population. Arch Gen Psychiatry, 60, 3947.Google Scholar
Okuda, A, Suzuki, T, Kishi, T, et al. (2010). Duration of untreated illness and antidepressant fluvoxamine response in major depressive disorder. Psychiatry Clin Neurosci, 64(3), 268273. doi:10.1111/j.1440-1819.2010.02091.x.Google Scholar
Pacchiarotti, I, Bond, DJ, Baldessarini, RJ, et al. (2013). The International Society for Bipolar Disorders (ISBD) task force report on antidepressant use in bipolar disorders. Am J Psychiatry, 170(11), 12491262. doi:10.1176/appi.ajp.2013.13020185.Google Scholar
Papakostas, GI, Fava, M, Thase, ME (2008). Treatment of SSRI-resistant depression: a meta-analysis comparing within- versus across-class switches. Biol Psychiatry, 63(7), 699704.Google Scholar
Parker, G (2000). Classifying depression: should paradigms lost be regained? Am J Psychiatry, 157, 11951203.Google Scholar
Partonen, T, Lonnqvist, J (1996). Moclobemide and fluoxetine in treatment of seasonal affective disorder. J Affect Disord, 41, 9399.Google Scholar
Paykel, ES, Tanner, J (1976). Life events, depressive relapse and maintenance treatment. Psychol Med, 6, 481485.Google Scholar
Paykel, ES, Ramana, R, Cooper, Z, et al. (1995). Residual symptoms after partial remission: an important outcome in depression. Psychol Med, 25, 11711180.Google Scholar
Perahia, DG, Wang, F, Mallinckrodt, CH, Walker, DJ, Detke, MJ (2006). Duloxetine in the treatment of major depressive disorder: a placebo- and paroxetine-controlled trial. Eur Psychiatry, 21, 367378.Google Scholar
Perahia, DG, Pritchett, YL, Kajdasz, DK, et al. (2008). A randomized, double-blind comparison of duloxetine and venlafaxine in the treatment of patients with major depressive disorder. J Psychiatr Res, 42, 2234.Google Scholar
Petersen, I, Gilbert, RE, Evans, SJ, Man, SL, Nazareth, I (2011). Pregnancy as a major determinant for discontinuation of antidepressants: an analysis of data from The Health Improvement Network. J Clin Psychiatry, 72(7), 979985. doi:10.4088/JCP.10m06090blu.Google Scholar
Pope, HG Jr, Cohane, GH, Kanayama, G, Siegel, AJ, Hudson, JI (2003). Testosterone gel supplementation for men with refractory depression: a randomized, placebo-controlled trial. Am J Psychiatry, 160(1), 105111.Google Scholar
Pope, HG, Amiaz, R, Brennan, BP, et al. (2010). Parallel-group placebo-controlled trial of testosterone gel in men with major depressive disorder displaying an incomplete response to standard antidepressant treatment. J Clin Psychopharmacol, 30(2), 126134. doi:10.1097/JCP.0b013e3181d207ca.Google Scholar
Posternak, MA, Zimmerman, M (2005). Is there a delay in the antidepressant effect? A meta-analysis. J Clin Psychiatry, 66, 148158.Google Scholar
Posternak, MA, Zimmerman, M (2007). Therapeutic effect of follow-up assessments on antidepressant and placebo response rates in antidepressant efficacy trials: meta-analysis. Br J Psychiatry, 190, 287292.Google Scholar
Posternak, MA, Solomon, DA, Leon, AC, et al. (2006). The naturalistic course of unipolar major depression in the absence of somatic therapy. J Nerv Ment Dis, 194, 324329.Google Scholar
Quitkin, FM, Petkova, E, McGrath, PJ, et al. (2003). When should a trial of fluoxetine for major depression be declared failed? Am J Psychiatry, 160, 734740.Google Scholar
Raison, CL, Rutherford, RE, Woolwine, BJ, et al. (2013). A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA Psychiatry, 70(1), 3141. doi:10.1001/2013.jamapsychiatry.4.Google Scholar
Ramana, R, Paykel, ES, Cooper, Z, et al. (1995). Remission and relapse in major depression: a two-year prospective follow-up study. Psychol Med, 25, 11611170.Google Scholar
Reimherr, FW, Amsterdam, JD, Quitkin, FM, et al. (1998). Optimal length of continuation therapy in depression: a prospective assessment during long-term fluoxetine treatment. Am J Psychiatry, 155, 12471253.Google Scholar
Reimherr, FW, Strong, RE, Marchant, BK, Hedges, DW, Wender, PH (2001). Factors affecting return of symptoms 1 year after treatment in a 62-week controlled study of fluoxetine in major depression. J Clin Psychiatry, 62(Suppl 22), 1623.Google Scholar
Reis, M, Aamo, T, Spigset, O, Ahlner, J (2009). Serum concentrations of antidepressant drugs in a naturalistic setting: compilation based on a large therapeutic drug monitoring database. Ther Drug Monit, 31(1), 4256. doi:10.1097/FTD.0b013e31819114ea.Google Scholar
Reynolds, CF, III, Perel, JM, Frank, E, et al. (1999). Three-year outcomes of maintenance nortriptyline treatment in late-life depression: a study of two fixed plasma levels. Am J Psychiatry, 156, 11771181.Google Scholar
Reynolds, CF, III, Dew, MA, Pollock, BG, et al. (2006). Maintenance treatment of major depression in old age. N Engl J Med, 354, 11301138.Google Scholar
Robinson, P, Katon, W, Von Korff, M, et al. (1997). The education of depressed primary care patients: what do patients think of interactive booklets and a video?J Fam Pract, 44, 562571.Google Scholar
Rojo, JE, Ros, S, Aguera, L, de la Gandara, J, de Pedro, JM (2005). Combined antidepressants: clinical experience. Acta Psychiatr Scand Suppl, 428, 2531.Google Scholar
Ronalds, C, Creed, F, Stone, K, Webb, S, Tomenson, B (1997). The outcome of anxiety and depressive disorders in general practice. Br J Psychiatry, 171, 427433.Google Scholar
Rouillon, F, Serrurier, D, Miller, HD, Gerard, MJ (1991). Prophylactic efficacy of maprotiline on unipolar depression relapse. J Clin Psychiatry, 52, 423431.Google Scholar
Rudolph, RL, Fabre, LF, Feighner, JP, et al. (1998). A randomized, placebo-controlled, dose-response trial of venlafaxine hydrochloride in the treatment of major depression. J Clin Psychiatry, 59, 116122.Google Scholar
Ruhe, HG, Huyser, J, Swinkels, JA, Schene, AH (2006). Switching antidepressants after a first selective serotonin reuptake inhibitor in major depressive disorder: a systematic review. J Clin Psychiatry, 67, 18361855.Google Scholar
Rush, AJ, Trivedi, MH, Wisniewski, SR, et al. (2006a). Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry, 163, 19051917.Google Scholar
Rush, AJ, Trivedi, MH, Wisniewski, SR, et al. (2006b). Bupropion-SR, sertraline or venlafaxine-XR after failure of SSRIs for depression. N Engl J Med, 354, 12311242.Google Scholar
Rush, AJ, Trivedi, MH, Stewart, JW, et al. (2011). Combining medications to enhance depression outcomes (CO-MED): acute and long-term outcomes of a single-blind randomized study. Am J Psychiatry, 168, 689701.Google Scholar
Sachs, GS, Nierenberg, AA, Calabrese, JR, et al. (2007). Effectiveness of adjunctive antidepressant treatment for bipolar depression. N Engl J Med, 356(17), 17111722.Google Scholar
Sansone, RA, Sansone, LA (2012). Antidepressant adherence: are patients taking their medications? Innov Clin Neurosci, 9(5–6), 4146.Google Scholar
Schindler, F, Anghelescu, IG (2007). Lithium versus lamotrigine augmentation in treatment resistant unipolar depression: a randomized, open-label study. Int Clin Psychopharmacol, 22, 179182.Google Scholar
Schmidt, ME, Fava, M, Zhang, S, et al. (2002). Treatment approaches to major depressive disorder relapse. Part 1: dose increase. Psychother Psychosom, 71, 9094.Google Scholar
Schueler, Y-B, Koesters, M, Wieseler, B, et al. (2011). A systematic review of duloxetine and venlafaxine in major depression, including unpublished data. Acta Psychiatr Scand, 123, 247265.Google Scholar
Schweizer, E, Rynn, M, Mandos, LA, et al. (2001). The antidepressant effect of sertraline is not enhanced by dose titration: results from an outpatient clinical trial. Int Clin Psychopharmacol, 16, 137143.CrossRefGoogle Scholar
Servier Laboratories Limited (2012). SPC for Valdoxan.Google Scholar
Simon, GE, Savarino, J (2007). Suicide attempts among patients starting depression treatment with medications or psychotherapy. Am J Psychiatry, 164, 10291034.Google Scholar
Simon, GE, Savarino, J, Operskalski, B, Wang, PS (2006). Suicide risk during antidepressant treatment. Am J Psychiatry, 163, 4147.Google Scholar
Solomon, DA, Keller, MB, Leon, AC, et al. (2000). Multiple recurrences of major depressive disorder. Am J Psychiatry, 157, 229233.Google Scholar
Solomon, DA, Leon, AC, Mueller, TI, et al. (2005). Tachyphylaxis in unipolar major depressive disorder. J Clin Psychiatry, 66, 283290.Google Scholar
Spielmans, GI, Berman, MI, Linardatos, E, et al. (2013). Adjunctive atypical antipsychotic treatment for major depressive disorder: a meta-analysis of depression, quality of life, and safety outcomes. PLoS Med, 10(3), e1001403.Google Scholar
Spina, E, Santoro, V, D’Arrigo, C (2008). Clinically relevant pharmacokinetic drug interactions with second-generation antidepressants: an update. Clin Ther, 30(7), 12061227.Google Scholar
Stassen, HH, Angst, J, Delini-Stula, A (1996). Delayed onset of action of antidepressant drugs? Survey of results of Zurich meta-analyses. Pharmacopsychiatry, 29, 8796.Google Scholar
Strawbridge, R, Carter, B, Marwood, L, et al. (2019). Augmentation therapies for treatment-resistant depression: systematic review and meta-analysis. Br J Psychiatry, 214(1), 4251. doi:10.1192/bjp.2018.233.Google Scholar
Suppes, T, Silva, R, Cucchiaro, J, et al. (2016). Lurasidone for the treatment of major depressive disorder with mixed features: a randomized, double-blind, placebo-controlled study. Am J Psychiatry, 173, 400407.Google Scholar
Szegedi, A, Muller, MJ, Anghelescu, I, et al. (2003). Early improvement under mirtazapine and paroxetine predicts later stable response and remission with high sensitivity in patients with major depression. J Clin Psychiatry, 64, 413420.Google Scholar
Targownik, LE, Bolton, JM, Metge, CJ, Leung, S, Sareen, J (2009). Selective serotonin reuptake inhibitors are associated with a modest increase in the risk of upper gastrointestinal bleeding. Am J Gastroenterol, 104(6), 14751482. doi:10.1038/ajg.2009.128.Google Scholar
Taylor, D, Cornelius, V, Smith, L, Young, AH (2014). Comparative efficacy and acceptability of drug treatments for bipolar depression: a multiple-treatments meta-analysis. Acta Psychiatr Scand, 130(6), 452469. doi:10.1111/acps.12343.Google Scholar
Taylor, D, Barnes, TRE, Young, A (2018). The Maudsley Prescribing Guidelines in Psychiatry, 13th ed. Wiley-Blackwell.Google Scholar
Taylor, MJ, Freemantle, N, Geddes, JR, Bhagwagar, Z (2006). Early onset of selective serotonin reuptake inhibitor antidepressant action: systematic review and meta-analysis. Arch Gen Psychiatry, 63, 12171223.Google Scholar
Taylor, MJ, Rudkin, L, Bullemor-Day, P, et al. (2013). Strategies for managing sexual dysfunction induced by antidepressant medication. Cochrane Database Syst Rev, (5), CD003382. doi:10.1002/14651858.CD003382.pub3.Google Scholar
Thanacoody, HK, Thomas, SH (2005). Tricyclic antidepressant poisoning: cardiovascular toxicity. Toxicol Rev, 24, 205214.Google Scholar
Thase, ME (2006). Preventing relapse and recurrence of depression: a brief review of therapeutic options. CNS Spectr, 11, 1221.Google Scholar
Thase, ME, Trivedi, MH, Rush, AJ (1995). MAOIs in the contemporary treatment of depression. Neuropsychopharmacology, 12, 185219.Google Scholar
Thase, ME, Shelton, RC, Khan, A (2006). Treatment with venlafaxine extended release after SSRI nonresponse or intolerance: a randomized comparison of standard- and higher-dosing strategies. J Clin Psychopharmacol, 26, 250258.Google Scholar
Thase, ME, Larsen, KG, Reines, E, Kennedy, SH (2013). The cardiovascular safety profile of escitalopram. Eur Neuropsychopharmacol, 23(11), 13911400. doi:10.1016/j.euroneuro.2013.05.011.Google Scholar
Thase, ME, Mahableshwarkar, AR, Dragheim, M, Loft, H, Vieta, E (2016). A meta-analysis of randomized, placebo-controlled trials of vortioxetine for the treatment of major depressive disorder in adults. Eur Neuropsychopharmacol, 26, 979993.Google Scholar
Tint, A, Haddad, PM, Anderson, IM (2008). The effect of rate of antidepressant tapering on the incidence of discontinuation symptoms: a randomised study. J Psychopharmacol, 22(3), 330332.Google Scholar
Tomaszewska, W, Peselow, ED, Barouche, F, Fieve, RR (1996). Antecedent life events, social supports and response to antidepressants in depressed patients. Acta Psychiatr Scand, 94, 352357.Google Scholar
Trivedi, MH, Fava, M, Wisniewski, SR, et al. (2006a). Medication augmentation after the failure of SSRIs for depression. N Engl J Med, 354, 12431252.Google Scholar
Trivedi, MH, Rush, AJ, Wisniewski, SR, et al. (2006b). Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry, 163, 2840.Google Scholar
Uher, R, Maier, W, Hauser, J, et al. (2009). Differential efficacy of escitalopram and nortriptyline on dimensional measures of depression. Br J Psychiatry, 194, 252259. doi:10.1192/bjp.bp.108.057554.Google Scholar
Vallejo, J, Gasto, C, Catalan, R, Bulbena, A, Menchon, JM (1991). Predictors of antidepressant treatment outcome in melancholia: psychosocial, clinical and biological indicators. J Affect Disord, 21, 151162.Google Scholar
Vergouwen, AC, Bakker, A, Katon, WJ, Verheij, TJ, Koerselman, F (2003). Improving adherence to antidepressants: a systematic review of interventions. J Clin Psychiatry, 64, 14151420.Google Scholar
Vieta, E, Bauer, M, Montgomery, S, et al. (2013). Pooled analysis of sustained response rates for extended release quetiapine fumarate as monotherapy or adjunct to antidepressant therapy in patients with major depressive disorder. J Affect Disord, 150(2), 639643. doi:10.1016/j.jad.2013.01.052.Google Scholar
Walsh, BT, Seidman, SN, Sysko, R, Gould, M (2002). Placebo response in studies of major depression: variable, substantial, and growing. JAMA, 287, 18401847.Google Scholar
Walters, G, Reynolds, CF, III, Mulsant, BH, Pollock, BG (1999). Continuation and maintenance pharmacotherapy in geriatric depression: an open-trial comparison of paroxetine and nortriptyline in patients older than 70 years. J Clin Psychiatry, 60(Suppl 20), 2125.Google Scholar
Walther, A, Breidenstein, J, Miller, R (2019). Association of testosterone treatment with alleviation of depressive symptoms in men: a systematic review and meta-analysis. JAMA Psychiatry, 76(1), 3140. doi:10.1001/jamapsychiatry.2018.2734.Google Scholar
Warden, D, Rush, AJ, Trivedi, MH, Fava, M, Wisniewski, SR (2007). The STAR*D Project results: a comprehensive review of findings. Curr Psychiatry Rep, 9(6), 449459.Google Scholar
Wijkstra, J, Lijmer, J, Balk, JF, Geddes, JR., Nolen, WA (2006). Pharmacological treatment for unipolar psychotic depression: systematic review and meta-analysis. Br J Psychiatry, 188(5), 410415.Google Scholar
Wohlreich, MM, Mallinckrodt, CH, Watkin, JG, et al. (2005). Immediate switching of antidepressant therapy: results from a clinical trial of duloxetine. Ann Clin Psychiatry, 17, 259268.Google Scholar
Yildiz, A, Pauler, DK, Sachs, GS (2004). Rates of study completion with single versus split daily dosing of antidepressants: a meta-analysis. J Affect Disord, 78, 157162.Google Scholar
Yyldyz, A, Sachs, GS (2001). Administration of antidepressants. Single versus split dosing: a meta-analysis. J Affect Disord, 66, 199206.Google Scholar
Zarrouf, FA, Artz, S, Griffith, J, Sirbu, C, Kommor, M (2009). Testosterone and depression: systematic review and meta-analysis. J Psychiatr Pract, 15(4), 289305. doi:10.1097/01.pra.0000358315.88931.fc.Google Scholar
Zheng, W, Li, X-H, Zhu, X-M, et al. (2019). Adjunctive ketamine and electroconvulsive therapy for major depressive disorder: a meta-analysis of randomized controlled trials. J Affective Disord, 250, 123131.Google Scholar
Zhou, X, Ravindran, AV, Qin, B, et al. (2015). Comparative efficacy, acceptability, and tolerability of augmentation agents in treatment-resistant depression: systematic review and network meta-analysis. Clin Psychiatry, 76(4), e487e498. doi:10.4088/JCP.14r09204.Google Scholar
Zivin, K, Pfeiffer, PN, Bohnert, AS, et al. (2013). Evaluation of the FDA warning against prescribing citalopram at doses exceeding 40 mg. Am J Psychiatry, 170(6), 642650. doi:10.1176/appi.ajp.2013.12030408.Google Scholar

References

Argolo, FC, Cavalcanti-Ribeiro, P, Netto, LR, Quarantini, LC (2015). Prevention of posttraumatic stress disorder with propranolol: a meta-analytic review. J Psychosom Res, 79, 8993.Google Scholar
Baldwin, DS, Aitchison, KBA, Curran, HV, et al. (2013). Benzodiazepines: risks and benefits. A reconsideration. J Psychopharmacol, 27, 967971.Google Scholar
Baldwin, DS, Anderson, IM, Nutt, DJ, et al. (2014). Evidence-based pharmacological treatment of anxiety disorders, post-traumatic stress disorder and obsessive-compulsive disorder: a revision of the 2005 guidelines from the British Association for Psychopharmacology. J Psychopharmacol, 28, 403439.Google Scholar
Baldwin, DS, den Boer, JA, Lyndon, G, et al. (2015). Efficacy and safety of pregabalin in generalised anxiety disorder: a critical review of the literature. J Psychopharmacol, 29, 10471060.CrossRefGoogle ScholarPubMed
Bandelow, B, Reitt, M, Röver, C, et al. (2015). Efficacy of treatments for anxiety disorders: a meta-analysis. Int Clin Psychopharmacol, 30, 183192.Google Scholar
Bandelow, B, Baldwin, D, Abelli, M, et al. (2017). Biological markers for anxiety disorders, OCD and PTSD: a consensus statement. Part II: Neurochemistry, neurophysiology and neurocognition. World J Biol Psychiatry, 18, 162214.Google Scholar
Barker, MJ, Greenwood, KM, Jackson, M, Crowe, SF (2004). Persistence of cognitive effects after withdrawal from long-term benzodiazepine use: a meta-analysis. Arch Clin Neuropsychol, 19, 437454.Google Scholar
Batelaan, NM, Bosman, RC, Muntingh, A, et al. (2017). Risk of relapse after antidepressant discontinuation in anxiety disorders, obsessive-compulsive disorder, and post-traumatic stress disorder: systematic review and meta-analysis of relapse prevention trials. BMJ, 358, j3927.Google Scholar
Billioti de Gage, S, Pariente, A, Bégaud, B (2015). Is there really a link between benzodiazepine use and the risk of dementia? Expert Opin Drug Saf, 14, 733747.Google Scholar
Bonnet, U, Scherbaum, N (2017). How addictive are gabapentin and pregabalin? A systematic review. Eur Neuropsychopharmacol, 27, 11851215.Google Scholar
Chagraoui, A, Skiba, M, Thuillez, C, Thibaut, F (2016). To what extent is it possible to dissociate the anxiolytic and sedative/hypnotic properties of GABAA receptors modulators? Prog Neuropsychopharmacol Biol Psychiatry, 71, 189202.Google Scholar
Chessick, CA, Allen, MH, Thase, ME, et al. (2006). Azapirones for generalized anxiety disorder. Cochrane Database Syst Rev, (3), CD006115.Google Scholar
Corchs, F, Nutt, DJ, Hince, DA, et al. (2015). Evidence for serotonin function as a neurochemical difference between fear and anxiety disorders in humans? J Psychopharmacol, 29, 10611069.Google Scholar
Cowen, PJ, Anderson, IM, Grahame-Smith, DG (1990). Neuroendocrine effects of azapirones. J Clin Psychopharmacol, 10, 21S25S.Google Scholar
Culpepper, L, Wingertzahn, MA (2015). Over-the-counter agents for the treatment of occasional disturbed sleep or transient insomnia: a systematic review of efficacy and safety. Prim Care Companion CNS Disord, 17, 10.4088/PCC.15r01798.Google Scholar
Deakin, JFW (2013). The origins of ‘5-HT and mechanisms of defence’ by Deakin and Graeff: a personal perspective. J Psychopharmacol, 27, 10841089.Google Scholar
Dold, M, Aigner, M, Lanzenberger, R, Kasper, S (2015). Antipsychotic augmentation of serotonin reuptake inhibitors in treatment-resistant obsessive-compulsive disorder: an update meta-analysis of double-blind, randomized, placebo-controlled trials. Int J Neuropsychopharmacol, 18, pii: pyv047. doi:10.1093/ijnp/pyv047.Google Scholar
Drover, DR (2004). Comparative pharmacokinetics and pharmacodynamics of short-acting hypnosedatives. Clin Pharmacokinet, 43, 227238.Google Scholar
Ducasse, D, Boyer, L, Michel, P, et al. (2014). D2 and D3 dopamine receptor affinity predicts effectiveness of antipsychotic drugs in obsessive-compulsive disorders: a metaregression analysis. Psychopharmacology (Berl), 231, 37653770.Google Scholar
Dündar, Y, Dodd, S, Strobl, J, et al. (2004). Comparative efficacy of newer hypnotic drugs for the short-term management of insomnia: a systematic review and meta-analysis. Hum Psychopharmacol, 19, 305322.Google Scholar
Fleminger, S, Greenwood, RRJ, Oliver, DL (2006). Pharmacological management for agitation and aggression in people with acquired brain injury. Cochrane Database Syst Rev, (4), CD003299.Google Scholar
Freynhagen, R, Backonja, M, Schug, S, et al. (2016). Pregabalin for the treatment of drug and alcohol withdrawal symptoms: a comprehensive review. CNS Drugs, 30, 11911200.Google Scholar
Goodwin, GM, Emsley, R, Rembry, S, et al. (2009). Agomelatine prevents relapse in patients with major depressive disorder without evidence of a discontinuation syndrome: a 24-week randomized, double-blind, placebo-controlled trial. J Clin Psychiatry, 70, 11281137.Google Scholar
Haddad, PM, Anderson, IM (2007). Recognising and managing antidepressant discontinuation symptoms. Adv Psychiatr Treat, 13, 447457.Google Scholar
Hajak, G, Müller, WE, Wittchen, HU, Pittrow, D, Kirch, W (2003). Abuse and dependence potential for the non-benzodiazepine hypnotics zolpidem and zopiclone: a review of case reports and epidemiological data. Addiction, 98, 13711378.Google Scholar
Kalueff, AV, Nutt, DJ (2007). Role of GABA in anxiety and depression. Depress Anxiety, 24, 495517.Google Scholar
Kishi, T, Meltzer, HY, Matsuda, Y, Iwata, N (2013). Azapirone 5-HT1A receptor partial agonist treatment for major depressive disorder: systematic review and meta-analysis. Psychol Med, 44, 22552269.Google Scholar
Kishi, T, Matsunaga, S, Iwata, N (2015). Suvorexant for primary insomnia: a systematic review and meta-analysis of randomized placebo-controlled trials. PLoS One, 10, e0136910.Google Scholar
Kuriyama, A, Honda, M, Hayashino, Y (2014). Ramelteon for the treatment of insomnia in adults: a systematic review and meta-analysis. Sleep Med, 15, 385392.Google Scholar
Loane, C, Politis, M (2012). Buspirone: what is it all about? Brain Res, 1461, 111118.Google Scholar
Maneeton, N, Maneeton, B, Woottiluk, P, et al. (2016). Quetiapine monotherapy in acute treatment of generalized anxiety disorder: a systematic review and meta-analysis of randomized controlled trials. Drug Des Devel Ther, 10, 259276.Google Scholar
Micó, J-A, Prieto, R (2012). Elucidating the mechanism of action of pregabalin. CNS Drugs, 26, 637648.Google Scholar
Miller, CH, Fleischhacker, WW (2000). Managing antipsychotic-induced acute and chronic akathisia. Drug Saf, 22, 7381.Google Scholar
Muscatello, MR, Spina, E, Bandelow, B, Baldwin, DS (2012). Clinically relevant drug interactions in anxiety disorders. Hum Psychopharmacol, 27, 239253.Google Scholar
Peterlik, D, Flor, PJ, Uschold-Schmidt, N (2016). The emerging role of metabotropic glutamate receptors in the pathophysiology of chronic stress-related disorders. Curr Neuropharmacol, 14, 514539.Google Scholar
Pignon, B, Tezenas du Montcel, C, Carton, L, Pelissolo, A (2017). The place of antipsychotics in the therapy of anxiety disorders and obsessive-compulsive disorders. Curr Psychiatry Rep, 19, 103.Google Scholar
Pitsikas, N (2014). The metabotropic glutamate receptors: potential drug targets for the treatment of anxiety disorders? Eur J Pharmacol, 723, 181184.Google Scholar
Schjerning, O, Rosenzweig, M, Pottegård, A, Damkier, P, Nielsen, J (2016). Abuse potential of pregabalin. CNS Drugs, 30, 925.Google Scholar
Sieghart, W, Sperk, G (2002). Subunit composition, distribution and function of GABA-A receptor subtypes. Curr Top Med Chem, 2, 795816.Google Scholar
Sigel, E, Steinmann, ME (2012). Structure, function and modulation of GABAA receptor. J Biol Chem, 287, 4022440231.Google Scholar
Soyka, M (2017). Treatment of benzodiazepine dependence. N Engl J Med, 376, 11471157.Google Scholar
Steenen, SA, van Wijk, AJ, van der Heijden, GJ, et al. (2016). Propranolol for the treatment of anxiety disorders: systematic review and meta-analysis. J Psychopharmacol, 30, 128139.Google Scholar
Stein, DJ, Ahokas, AA, de Bodinat, C (2008). Efficacy of agomelatine in generalized anxiety disorder: a randomized, double-blind, placebo-controlled study. J Clin Psychopharmacol, 28, 561566.Google Scholar
Stein, DJ, Ahokas, A, Albarran, C, et al. (2012). Agomelatine prevents relapse in generalized anxiety disorder: a 6-month randomized, double-blind, placebo-controlled discontinuation study. J Clin Psychiatry, 73, 10021008.Google Scholar
Sutton, JA, Clauss, RP (2017). A review of the evidence of zolpidem efficacy in neurological disability after brain damage due to stroke, trauma and hypoxia: a justification of further clinical trials. Brain Inj, 31, 10191027.Google Scholar
Turner, P, Granville-Grossman, KL, Smart, JV (1965). Effect of adrenergic receptor blockade on the tachycardia of thyrotoxicosis and anxiety state. Lancet, 286, 13161318.Google Scholar
Walsh, JK, Krystal, AD, Amato, DA, et al. (2007). Nightly treatment of primary insomnia with eszopiclone for six months: effect on sleep, quality of life, and work limitations. Sleep, 30, 959968.CrossRefGoogle ScholarPubMed
Whale, R, Terao, T, Cowen, P, Freemantle, C, Geddes, G (2010). Pindolol augmentation of serotonin reuptake inhibitors for the treatment of depressive disorder: a systematic review. J Psychopharmacol, 24, 513520.Google Scholar
Wilson, SJ, Nutt, DJ, Alford, C, et al. (2010). British Association for Psychopharmacology consensus statement on evidence-based treatment of insomnia, parasomnias and circadian rhythm disorders. J Psychopharmacol, 24, 15771601.Google Scholar
Yeung, W-F, Chung, K-F, Yung, K-P, Ng, TH-Y (2015). Doxepin for insomnia: a systematic review of randomized placebo-controlled trials. Sleep Med Rev, 19, 7583.Google Scholar

References

Adams, CE, Fenton, MK, Quraishi, S, David, AS (2001). Systematic meta-review of depot antipsychotic drugs for people with schizophrenia. Br J Psychiatry, 179, 290299.Google Scholar
Arranz, MJ, Rivera, M, Munro, JC (2011). Pharmacogenetics of response to antipsychotics in patients with schizophrenia. CNS Drugs, 25(11), 933939.Google Scholar
Ashok, AH, Marques, TR, Jauhar, S, et al. (2017). The dopamine hypothesis of bipolar affective disorder: the state of the art and implications for treatment. Mol Psychiatry, 22(5), 666679.Google Scholar
Bagnall, AM, Jones, L, Ginnelly, L, et al. (2003). A systematic review of atypical antipsychotic drugs in schizophrenia. Health Technol Assess, 7(13), 1193.Google Scholar
Beck, K, McCutcheon, R, Bloomfield, MAP, et al. (2014). The practical management of refractory schizophrenia – the Maudsley Treatment Review and Assessment Team service approach. Acta Psychiatr Scand, 130(6), 427438.Google Scholar
Beresford, R, Ward, A (1987). Haloperidol decanoate. A preliminary review of its pharmacodynamic and pharmacokinetic properties and therapeutic use in psychosis. Drugs, 33(1), 3149.Google Scholar
Bleakley, S (2012). Identifying and reducing the risk of antipsychotic drug interactions. Prog Neurol Psychiatry, 16(2), 2024.Google Scholar
Bolden, C, Cusack, B, Richelson, E (1992). Antagonism by antimuscarinic and neuroleptic compounds at the five cloned human muscarinic cholinergic receptors expressed in Chinese hamster ovary cells. J Pharmacol Exp Ther, 260(2), 576580.Google Scholar
Brugger, SP, Howes, OD (2017). Heterogeneity and homogeneity of regional brain structure in schizophrenia: a meta-analysis. JAMA Psychiatry, 74(11), 11041111.Google Scholar
Buckley, PF, Schooler, NR, Goff, DC, et al. (2016). Comparison of injectable and oral antipsychotics in relapse rates in a pragmatic 30-month schizophrenia relapse prevention study. Psychiatr Serv, 67(12), 13701372.Google Scholar
Bymaster, FP, Calligaro, DO, Falcone, JF, et al. (1996). Radioreceptor binding profile of the atypical antipsychotic olanzapine. Neuropsychopharmacology, 14, 8796.Google Scholar
Carbon, M, Kane, MJ, Leucht, S, Correll, CU (2018). Tardive dyskinesia risk with first- and second-generation antipsychotics in comparative randomized controlled trials: a meta-analysis. World Psychiatry, 17(3), 330340. doi:10.1002/wps.20579.Google Scholar
Carlsson, A, Lindqvist, M (1963). Effect of chlorpromazine or haloperidol on formation of 3-methoxytyramine and normetanephrine in mouse brain. Acta Pharmacol Toxicol (Copenh), 20(2), 140144.Google Scholar
Cipriani, A, Barbui, C, Salanti, G, et al. (2011). Comparative efficacy and acceptability of antimanic drugs in acute mania: a multiple-treatments meta-analysis. Lancet, 378(9799), 13061315.Google Scholar
Correll, CU (2010). From receptor pharmacology to improved outcomes: individualising the selection, dosing, and switching of antipsychotics. Eur Psychiatry, 25(Suppl 2), S12S21. doi:10.1016/S0924-9338(10)71701-6.Google Scholar
Correll, CU, Citrome, L, Haddad, PM, et al. (2016). The use of long-acting injectable antipsychotics in schizophrenia: evaluating the evidence. J Clin Psychiatry, 77(Suppl 3), 124.Google Scholar
Creese, I, Burt, D, Snyder, S (1976). Dopamine receptor binding predicts clinical and pharmacological potencies of antischizophrenic drugs. Science, 192(4238), 481483.Google Scholar
Davidson, M, Saoud, J, Staner, C, et al. (2017). Efficacy and safety of MIN-101: a 12-week randomized, double-blind, placebo-controlled trial of a new drug in development for the treatment of negative symptoms in schizophrenia. Am J Psychiatry, 174(12), 11951202.Google Scholar
Davies, DL, Shepherd, M (1955). Reserpine in the treatment of anxious and depressed patients. Lancet, 269(6881), 117120.Google Scholar
Dold, M, Aigner, M, Lanzenberger, R, Kasper, S (2015). Antipsychotic augmentation of serotonin reuptake inhibitors in treatment-resistant obsessive-compulsive disorder: an update meta-analysis of double-blind, randomized, placebo-controlled trials. Int J Neuropsychopharmacol, 18(9), pyv047.Google Scholar
Douglas, IJ, Smeeth, L (2008). Exposure to antipsychotics and risk of stroke: self controlled case series study. BMJ, 337, a1227.Google Scholar
Dzahini, O, Singh, N, Taylor, D, Haddad, PM (2018). Antipsychotic drug use and pneumonia: systematic review and meta-analysis. J Psychopharmacol, 32(11), 11671181.Google Scholar
Emsley, R, Medori, R, Koen, L, et al. (2008). Long-acting injectable risperidone in the treatment of subjects with recent-onset psychosis: a preliminary study. J Clin Psychopharmacol, 28(2), 210213.Google Scholar
Fallon, P, Dursun, S, Deakin, B (2012). Drug-induced supersensitivity psychosis revisited: characteristics of relapse in treatment-compliant patients. Ther Adv Psychopharmacol, 2(1), 1322.Google Scholar
Frampton, JE (2010). Olanzapine long-acting injection: a review of its use in the treatment of schizophrenia. Drugs, 70(17), 22892313.Google Scholar
González-Blanco, L, Greenhalgh, AMD, Garcia-Rizo, C, et al. (2016). Prolactin concentrations in antipsychotic-naïve patients with schizophrenia and related disorders: a meta-analysis. Schizophr Res, 174(1), 156160.Google Scholar
Goodwin, GM, Haddad, PM, Ferrier, IN, et al. (2016). Evidence-based guidelines for treating bipolar disorder: revised third edition recommendations from the British Association for Psychopharmacology. J Psychopharmacol, 30(6), 495553.Google Scholar
Grace, AA, Gomes, FV (2019). The circuitry of dopamine system regulation and its disruption in schizophrenia: insights into treatment and prevention. Schizophr Bull, 45(1), 148157. doi:10.1093/schbul/sbx199.Google Scholar
Graff-Guerrero, A, Mizrahi, R, Agid, O, et al. (2009). The dopamine D2 receptors in high-affinity state and D3 receptors in schizophrenia: a clinical [11C]-(+)-PHNO PET study. Neuropsychopharmacology, 34(4), 10781086.Google Scholar
Graff-Guerrero, A, Mamo, D, Shammi, C, et al. (2013). The effect of antipsychotics on the high-affinity state of D2 and D3 receptors: a positron emission tomography study with [11c]-(+)-PHNO. Arch Gen Psychiatry, 66(6), 606615.Google Scholar
Grossman, I, Sullivan, PF, Walley, N, et al. (2008). Genetic determinants of variable metabolism have little impact on the clinical use of leading antipsychotics in the CATIE study. Genet Med, 10(10), 720729.Google Scholar
Haddad, PM, Anderson, IM (2002). Antipsychotic-related QTc prolongation, torsade de pointes and sudden death. Drugs, 62(11), 16491671.Google Scholar
Haddad, PM, Correll, CU (2018). The acute efficacy of antipsychotics in schizophrenia: a review of recent meta-analyses. Ther Adv Psychopharmacol, 8(11), 303318.Google Scholar
Haddad, PM, Wieck, A (2004). Antipsychotic-induced hyperprolactinaemia: mechanisms, clinical features and management. Drugs, 64, 22912314.Google Scholar
Hedges, D, Jeppson, K, Whitehead, P (2003). Antipsychotic medication and seizures: a review. Drugs Today (Barc), 39(7), 551557.Google Scholar
Heres, S, Hamann, J, Kissling, W, Leucht, S (2006). Attitudes of psychiatrists toward antipsychotic depot medication. J Clin Psychiatry, 67(12), 19481953.Google Scholar
Hirsch, SR, Gaind, R, Rohde, PD, Stevens, BC, Wing, JK (1973). Outpatient maintenance of chronic schizophrenic patients with long-acting fluphenazine: double-blind placebo. Br Med J, 1(5854), 633637.Google Scholar
Ho, BC, Andreasen, NC, Ziebell, S, Pierson, R, Magnotta, V (2011). Long-term antipsychotic treatment and brain volumes: a longitudinal study of first-episode schizophrenia. Arch Gen Psychiatry, 68(2), 128137.Google Scholar
Horvitz-Lennon, M, Mattke, S, Predmore, Z, Howes, OD (2017). The role of antipsychotic plasma levels in the treatment of schizophrenia. Am J Psychiatry, 174(5), 421426.Google Scholar
Howes, OD, Kapur, S (2014). A neurobiological hypothesis for the classification of schizophrenia: type a (hyperdopaminergic) and type b (normodopaminergic). Br J Psychiatry, 205, 13.Google Scholar
Howes, OD, McCutcheon, R. (2017). Inflammation and the neural diathesis-stress hypothesis of schizophrenia: a reconceptualization. Transl Psychiatry, 7(2), e1024. doi:10.1038/tp.2016.278.Google Scholar
Howes, OD, Kambeitz, J, Kim, E, et al. (2012). The nature of dopamine dysfunction in schizophrenia and what this means for treatment. Arch Gen Psychiatry, 69(8), 776786.Google Scholar
Howes, O, McCutcheon, R, Stone, J (2015). Glutamate and dopamine in schizophrenia: an update for the 21(st) century. J Psychopharmacol, 29(2), 97115.Google Scholar
Howes, OD, McCutcheon, R, Agid, O, et al. (2017a). Treatment-resistant schizophrenia: Treatment Response and Resistance in Psychosis (TRRIP) working group consensus guidelines on diagnosis and terminology. Am J Psychiatry, 174(3), 216229.Google Scholar
Howes, OD, McCutcheon, R, Owen, MJ, Murray, RM (2017b). The role of genes, stress, and dopamine in the development of schizophrenia. Biol Psychiatry, 81(1), 920.Google Scholar
Jauhar, S, Veronese, M, Rogdaki, M, et al. (2017). Regulation of dopaminergic function: an [18 F]-DOPA PET apomorphine challenge study in humans. Transl Psychiatry, 7(2), e1027. doi:10.1038/tp.2016.270.Google Scholar
Johnsen, E, Kroken, RA, Abaza, M, Olberg, H, Jørgensen, HA (2008). Antipsychotic-induced hyperprolactinemia: a cross-sectional survey. J Clin Psychopharmacol, 28(6), 686690. doi:10.1097/JCP.0b013e31818ba5d8.Google Scholar
Johnson, DAW (2009). Historical perspective on antipsychotic long-acting injections. Br J Psychiatry, 195(52), S7S12.Google Scholar
Kane, JM, Detke, HC, Naber, D, et al. (2010). Olanzapine long-acting injection: a 24-week, randomized, double-blind trial of maintenance treatment in patients with schizophrenia. Am J Psychiatry, 167(2), 181189.Google Scholar
Kapur, S (1998). A new framework for investigating antipsychotic action in humans: lessons from PET imaging. Mol Psychiatry, 3(2), 135140.Google Scholar
Kapur, S, Seeman, P (2000). Antipsychotic agents differ in how fast they come off the dopamine D2 receptors. Implications for atypical antipsychotic action. J Psychiatry Neurosci, 25(2), 161166.Google Scholar
Kapur, S, Seeman, P (2001). Does fast dissociation from the dopamine D2 receptor explain the action of atypical antipsychotics?: a new hypothesis. Am J Psychiatry, 158(3), 360369.Google Scholar
Kapur, S, Zipursky, RB, Remington, G, et al. (1998). 5-HT2 and D2 receptor occupancy of olanzapine in schizophrenia: a PET investigation. Am J Psychiatry, 155(7), 921928.Google Scholar
Kapur, S, Zipursky, RB, Remington, G (1999). Clinical and theoretical implications of 5-HT2 and D2 receptor occupancy of clozapine, risperidone, and olanzapine in schizophrenia. Am J Psychiatry, 156(2), 286293.Google Scholar
Kapur, S, Zipursky, R, Jones, C, Remington, G, Houle, S (2000). Relationship between dopamine D2 occupancy, clinical response, and side effects: a double-blind PET study of first-episode schizophrenia. Am J Psychiatry, 157(4), 514520.Google Scholar
Kebabian, JW, Calne, DB (1979). Multiple receptors for dopamine. Nature, 277(5692), 9396.Google Scholar
Kim, B, Lee, SH, Choi, TK, et al. (2008). Effectiveness of risperidone long-acting injection in first-episode schizophrenia: in naturalistic setting. Prog Neuropsychopharmacol Biol Psychiatry, 32(5), 12311235. doi:10.1016/j.pnpbp.2008.03.012.Google Scholar
King, C, Voruganti, LNP (2002). What’s in a name? The evolution of the nomenclature of antipsychotic drugs. J Psychiatry Neurosci, 27(3), 168175.Google Scholar
Kirson, NY, Weiden, PJ, Yermakov, S, et al. (2013). Efficacy and effectiveness of depot versus oral antipsychotics in schizophrenia: synthesizing results across different research designs. J Clin Psychiatry, 74(6), 568575.Google Scholar
Kisely, S, Sawyer, E, Robinson, G, Siskind, D (2015). A systematic review and meta-analysis of the effect of depot antipsychotic frequency on compliance and outcome. Schizophr Res, 166(1–3), 178186.Google Scholar
Kishimoto, T, Robenzadeh, A, Leucht, C, et al. (2014). Long-acting injectable vs oral antipsychotics for relapse prevention in schizophrenia: a meta-analysis of randomized trials. Schizophr Bull, 40(1), 192213.Google Scholar
Kwentus, J, Riesenberg, RA, Marandi, M, et al. (2012). Rapid acute treatment of agitation in patients with bipolar I disorder: a multicenter, randomized, placebo-controlled clinical trial with inhaled loxapine. Bipolar Disord, 14(1), 3140. doi:10.1111/j.1399-5618.2011.00975.x.Google Scholar
Laborit, H, Huguenard, P, Alluaume, R (1952). Un noveau stabilisateur végétatif (le 4560 RP). La Presse Médicale, 60, 206–208.Google Scholar
Lambert, M, Conus, P, Eide, P, et al. (2004). Impact of present and past antipsychotic side effects on attitude toward typical antipsychotic treatment and adherence. Eur Psychiatry, 19(7), 415422.Google Scholar
Lesem, MD, Tran-Johnson, TK, Riesenberg, RA, et al. (2011). Rapid acute treatment of agitation in individuals with schizophrenia: multicentre, randomised, placebo-controlled study of inhaled loxapine. Br J Psychiatry, 198(1), 5158. doi:10.1192/bjp.bp.110.081513.Google Scholar
Leucht, S, Pitschel-Walz, G, Abraham, D, Kissling, W (1999). Efficacy and extrapyramidal side-effects of the new antipsychotics olanzapine, quetiapine, risperidone, and sertindole compared to conventional antipsychotics and placebo. A meta-analysis of randomized controlled trials. Schizophr Res, 35(1), 5168.Google Scholar
Leucht, S, Busch, R, Kissling, W, Kane, JM (2007). Early prediction of antipsychotic nonresponse among patients with schizophrenia. J Clin Psychiatry, 68(3), 352360.Google Scholar
Leucht, S, Corves, C, Arbter, D, et al. (2009). Second-generation versus first-generation antipsychotic drugs for schizophrenia: a meta-analysis. Lancet, 373(9657), 3141.Google Scholar
Leucht, S, Hierl, S, Kissling, W, Dold, M, Davis, JM (2012a). Putting the efficacy of psychiatric and general medicine medication into perspective: review of meta-analyses. Br J Psychiatry, 200(2), 97106.Google Scholar
Leucht, S, Tardy, M, Komossa, K, et al. (2012b). Maintenance treatment with antipsychotic drugs for schizophrenia. Cochrane Database Syst Rev, (5), CD008016.Google Scholar
Leucht, S, Cipriani, A, Spineli, L, et al. (2013). Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. Lancet, 382(9896), 951962.Google Scholar
Leucht, S, Leucht, C, Huhn, M, et al. (2017). Sixty years of placebo-controlled antipsychotic drug trials in acute schizophrenia: systematic review, Bayesian meta-analysis, and meta-regression of efficacy predictors. Am J Psychiatry, 174(10), 927942.Google Scholar
Lieberman, JA, 3rd (2004). Managing anticholinergic side effects. Prim Care Companion J Clin Psychiatry, 6(Suppl 2), 2023.Google Scholar
Lindström, L, Lindström, E, Nilsson, M, Höistad, M (2017). Maintenance therapy with second generation antipsychotics for bipolar disorder – a systematic review and meta-analysis. J Affect Disord, 213, 138150. doi:10.1016/j.jad.2017.02.012.Google Scholar
Mallikaarjun, S, Kane, JM, Bricmont, P, et al. (2013). Pharmacokinetics, tolerability and safety of aripiprazole once-monthly in adult schizophrenia: an open-label, parallel-arm, multiple-dose study. Schizophr Res, 150(1), 281288.Google Scholar
Maneeton, N, Maneeton, B, Woottiluk, P, et al. (2016). Quetiapine monotherapy in acute treatment of generalized anxiety disorder: a systematic review and meta-analysis of randomized controlled trials. Drug Des Devel Ther, 10, 259276. doi:10.2147/DDDT.S89485.Google Scholar
Marder, SR, Hubbard, JW, Van Putten, T, Midha, KK (1989). Pharmacokinetics of long-acting injectable neuroleptic drugs: clinical implications. Psychopharmacology, 98(4), 433439.Google Scholar
Mauri, MC, Volonteri, LS, Colasanti, A, et al. (2007). Clinical pharmacokinetics of atypical antipsychotics: a critical review of the relationship between plasma concentrations and clinical response. Clin Pharmacokinet, 46(5), 359388.Google Scholar
McCutcheon, R, Beck, K, Bloomfield, MA, et al. (2015). Treatment resistant or resistant to treatment? Antipsychotic plasma levels in patients with poorly controlled psychotic symptoms. J Psychopharmacol, 29(8), 892897.Google Scholar
McCutcheon, R, Beck, K, D’Ambrosio, E, et al. (2017). Antipsychotic plasma levels in the assessment of poor treatment response in schizophrenia. Acta Psychiatr Scand, 137(1), 3946.Google Scholar
McCutcheon, R, Beck, K, Jauhar, S, Howes, OD (2018). Defining the locus of dopaminergic dysfunction in schizophrenia: a meta-analysis and test of the mesolimbic hypothesis. Schizophr Bull, 44(6), 13011311.Google Scholar
McCutcheon, RA, Abi-Dargham, A, Howes, OD (2019). Schizophrenia, dopamine and the striatum: from biology to symptoms. Trends Neurosci, 42(3), 205220.Google Scholar
Meltzer, HY, Matsubara, S, Lee, J-C (1989). Classification of typical and atypical antipsychotic drugs on the basis of dopamine. J Pharmacol Exp Ther, 251(1), 238246.Google Scholar
Minzenberg, MJ, Poole, JH, Benton, C, Vinogradov, S (2004). Association of anticholinergic load with impairment of complex attention and memory in schizophrenia. Am J Psychiatry, 161(1), 116124.Google Scholar
Murphy, BP, Chung, Y-C, Park, T-W, McGorry, PD (2006). Pharmacological treatment of primary negative symptoms in schizophrenia: a systematic review. Schizophr Res, 88(1–3), 525.Google Scholar
Murray, RM, Quattrone, D, Natesan, S, et al. (2016). Should psychiatrists be more cautious about the long-term prophylactic use of antipsychotics? Br J Psychiatry, 209(5), 361365.Google Scholar
Nyberg, S, Farde, L, Eriksson, L, Halldin, C, Eriksson, B (1993). 5-HT2 and D2 dopamine receptor occupancy in the living human brain. Psychopharmacology, 110(3), 265272.Google Scholar
Oehl, M, Hummer, M, Fleischhacker, WW (2000). Compliance with antipsychotic treatment. Acta Psychiatr Scand, 102, 8386.Google Scholar
Olianas, MC, Maullu, C, Onali, P (1997). Effects of clozapine on rat striatal muscarinic receptors coupled to inhibition of adenylyl cyclase activity and on the human cloned m4 receptor. Br J Pharmacol, 122(3), 401408.Google Scholar
Ortiz-Orendain, J, Castiello-de Obeso, S, Colunga-Lozano, LE, et al. (2017). Antipsychotic combinations for schizophrenia. Cochrane Database Syst Rev, (6), CD009005. doi:10.1002/14651858.CD009005.pub2.Google Scholar
Pantelis, C, Velakoulis, D, McGorry, PD, et al. (2003). Neuroanatomical abnormalities before and after onset of psychosis: a cross-sectional and longitudinal MRI comparison. Lancet, 361(9354), 281288.Google Scholar
Park, EJ, Amatya, S, Kim, MS, et al. (2013). Long-acting injectable formulations of antipsychotic drugs for the treatment of schizophrenia. Arch Pharm Res, 36(6), 651659.Google Scholar
Parsons, B, Allison, DB, Loebel, A, et al. (2009). Weight effects associated with antipsychotics: a comprehensive database analysis. Schizophr Res, 110(1–3), 103110.Google Scholar
Pelonero, AL, Levenson, JL, Pandurangi, AK (1998). Neuroleptic malignant syndrome: a review. Psychiatr Serv, 49(9), 11631172.Google Scholar
Perreault, ML, Hasbi, A, Alijaniaram, M, et al. (2010). The dopamine D1-D2 receptor heteromer localizes in dynorphin/enkephalin neurons: increased high affinity state following amphetamine and in schizophrenia. J Biol Chem, 285(47), 3662536634.Google Scholar
Ravyn, D, Ravyn, V, Lowney, R, Nasrallah, HA (2013). CYP450 pharmacogenetic treatment strategies for antipsychotics: a review of the evidence. Schizophr Res, 149(1–3), 114.Google Scholar
Ronaldson, KJ (2017). Cardiovascular disease in clozapine-treated patients: evidence, mechanisms and management. CNS Drugs, 31(9), 777795.Google Scholar
Rummel-Kluge, C, Komossa, K, Schwarz, S, et al. (2010). Head-to-head comparisons of metabolic side effects of second generation antipsychotics in the treatment of schizophrenia: a systematic review and meta-analysis. Schizophr Res, 123(2–3), 225233.Google Scholar
Sachs, GS, Nierenberg, AA, Calabrese, JR, et al. (2007). Effectiveness of adjunctive antidepressant treatment for bipolar depression. N Engl J Med, 356, 17111722. doi:10.1056/NEJMoa064135.Google Scholar
Samara, MT, Dold, M, Gianatsi, M, et al. (2016). Efficacy, acceptability, and tolerability of antipsychotics in treatment-resistant schizophrenia: a network meta-analysis. JAMA Psychiatry, 73(3), 199210.Google Scholar
Seeman, P (2001). Antipsychotic drugs, dopamine receptors, and schizophrenia. Clin Neurosci Res, 1(1–2), 5360.Google Scholar
Seeman, P (2011). All roads to schizophrenia lead to dopamine supersensitivity and elevated dopamine D2High receptors. CNS Neurosci Ther, 17(2), 118132.Google Scholar
Seeman, P (2012). Dopamine agonist radioligand binds to both D2High and D2Low receptors, explaining why alterations in D2High are not detected in human brain scans. Synapse, 66(1), 8893.Google Scholar
Seeman, P, Chau-Wong, M, Tedesco, J, Wong, K (1975). Brain receptors for antipsychotic drugs and dopamine: direct binding assays. Proc Natl Acad Sci U S A, 72(11), 43764380.Google Scholar
Sepede, G, De Berardis, D, Gambi, F, et al. (2006). Olanzapine augmentation in treatment-resistant panic disorder: a 12-week, fixed-dose, open-label trial. J Clin Psychopharmacol, 26(1), 4549.Google Scholar
Shen, WW (1999). A history of antipsychotic drug development. Compr Psychiatry, 40, 407414.Google Scholar
Siskind, D, McCartney, L, Goldschlager, R, Kisely, S (2016). Clozapine v. first- and second-generation antipsychotics in treatment-refractory schizophrenia: systematic review and meta-analysis. Br J Psychiatry, 209(5), 385392.Google Scholar
Slifstein, M, van de Giessen, E, Van Snellenberg, J, et al. (2015). Deficits in prefrontal cortical and extrastriatal dopamine release in schizophrenia: a positron emission tomographic functional magnetic resonance imaging study. JAMA Psychiatry, 72(4), 316324.Google Scholar
Soderberg, MM, Dahl, ML (2013). Pharmacogenetics of olanzapine metabolism. Pharmacogenomics, 14(11), 13191336.Google Scholar
Soldin, OP, Mattison, DR (2009). Sex differences in pharmacokinetics and pharmacodynamics. Clin Pharmacokinet, 48(3), 143157.Google Scholar
Spielmans, GI, Berman, MI, Linardatos, E, et al. (2013). Adjunctive atypical antipsychotic treatment for major depressive disorder: a meta-analysis of depression, quality of life, and safety outcomes. PLoS Med, 10(3), e1001403.Google Scholar
Spina, E, de Leon, J (2007). Metabolic drug interactions with newer antipsychotics: a comparative review. Basic Clin Pharmacol Toxicol, 100(1), 422.Google Scholar
Stein, G, Wilkinson, G (eds.) (2007). Seminars in General Adult Psychiatry, 2nd ed. London: Royal College of Psychiatrists.Google Scholar
Taylor, D (2009). Psychopharmacology and adverse effects of antipsychotic long-acting injections: a review. Br J Psychiatry, 195(52), S13S19.Google Scholar
Tiihonen, J, Haukka, J, Taylor, M, et al. (2011). A nationwide cohort study of oral and depot antipsychotics after first hospitalization for schizophrenia. Am J Psychiatry, 168(6), 603609.Google Scholar
Tiihonen, J, Mittendorfer-Rutz, E, Majak, M, et al. (2017). Real-world effectiveness of antipsychotic treatments in a nationwide cohort of 29823 patients with schizophrenia. JAMA Psychiatry, 74(7), 686693.Google Scholar
Veijola, J, Guo, JY, Moilanen, JS, et al. (2014). Longitudinal changes in total brain volume in schizophrenia: relation to symptom severity, cognition and antipsychotic medication. PLoS One, 9(7), e101689.Google Scholar
Weiden, PJ, Schooler, NR, Weedon, JC, et al. (2009). A randomized controlled trial of long-acting injectable risperidone vs continuation on oral atypical antipsychotics for first-episode schizophrenia patients: initial adherence outcome. J Clin Psychiatry, 70(10), 13971406. doi:10.4088/JCP.09m05284yel.Google Scholar
Weinberger, DR (1987). Implications of normal brain development for the pathogenesis of schizophrenia. Arch Gen Psychiatry, 44(7), 660669.Google Scholar
Wijkstra, J, Lijmer, J, Burger, H, et al. (2015). Pharmacological treatment for psychotic depression. Cochrane Database Syst Rev, (7), CD004044.Google Scholar
Wu, CS, Wang, SC, Yeh, IJ, Liu, SK (2016). Comparative risk of seizure with use of first- and second-generation antipsychotics in patients with schizophrenia and mood disorders. J Clin Psychiatry, 77(5), e573e579.Google Scholar
Young, SL, Taylor, M, Lawrie, SM (2015). ‘First do no harm.’ A systematic review of the prevalence and management of antipsychotic adverse effects. J Psychopharmacol, 29(4), 353362.Google Scholar
Zeng, XP, Le, F, Richelson, E (1997). Muscarinic m4 receptor activation by some atypical antipsychotic drugs. Eur J Pharmacol, 321(3), 349354.Google Scholar
Zhang, ZJ, Yao, ZJ, Liu, W, Fang, Q, Reynolds, GP (2004). Effects of antipsychotics on fat deposition and changes in leptin and insulin levels. Magnetic resonance imaging study of previously untreated people with schizophrenia. Br J Psychiatry, 184, 5862.Google Scholar
Zhu, Y, Li, C, Huhn, M, et al. (2017). How well do patients with a first episode of schizophrenia respond to antipsychotics: a systematic review and meta-analysis. Eur Neuropsychopharmacol, 27(9), 835844. doi:10.1016/j.euroneuro.2017.06.011.Google Scholar
Zohar, J, Stahl, S, Moller, H-J, et al. (2015). A review of the current nomenclature for psychotropic agents and an introduction to the Neuroscience-based Nomenclature. Eur Neuropsychopharmacol, 25(12), 23182325.Google Scholar

References

Agid, O, Arenovich, T, Sajeev, G, et al. (2011). An algorithm-based approach to first-episode schizophrenia: response rates over 3 prospective antipsychotic trials with a retrospective data analysis. J Clin Psychiatry, 72, 14391444.Google Scholar
Bagnall, AM, Jones, L, Ginnelly, L, et al. (2003). A systematic review of atypical antipsychotic drugs in schizophrenia. Health Technol Assess, 7(13), 1193.Google Scholar
Barber, S, Olotu, U, Corsi, M, Cipriani, A (2017). Clozapine combined with different antipsychotic drugs for treatment-resistant schizophrenia. Cochrane Database Syst Rev, (3), CD006324.Google Scholar
Barnes, TR; Schizophrenia Consensus Group of the British Association for Psychopharmacology (2011). Evidence-based guidelines for the pharmacological treatment of schizophrenia: recommendations from the British Association for Psychopharmacology. J Psychopharmacol, 25, 567620.Google Scholar
Barnes, TRE, Drake, RJ, Dunn, G, et al. (2013). The effect of prior treatment with long-acting injectable antipsychotic drugs on randomised, clinical trial treatment outcomes. Br J Psychiatry, 203(3), 215220.Google Scholar
Barnes, TRE, Dye, S, Ferrier, N, et al. (2014). Consensus Statement on High-Dose Antipsychotic Medication. College Report CR190. London: Royal College of Psychiatrists.Google Scholar
Beri, A, Boydell, J (2014). Clozapine in borderline personality disorder: a review of the evidence. Ann Clin Psychiatry, 26(2), 139144.Google Scholar
Bowskill, S, Couchman, L, MacCabe, JH, Flanagan, RJ (2012). Plasma clozapine and norclozapine in relation to prescribed dose and other factors in patients aged 65 years and over: data from a therapeutic drug monitoring service, 1996–2010. Hum Psychopharmacol, 27(3), 277283.Google Scholar
Brosda, J, Jantschak, F, Pertz, HH (2014). Alpha2-adrenoceptors are targets for antipsychotic drugs. Psychopharmacology, 231(5), 801812.Google Scholar
Chang, A, Fox, SH (2016). Psychosis in Parkinson’s disease: epidemiology, pathophysiology, and management. Drugs, 76(11), 10931118.Google Scholar
Chang, JS, Ha, KS, Young Lee, K, Sik Kim, Y, Min Ahn, Y (2006). The effects of long-term clozapine add-on therapy on the rehospitalization rate and the mood polarity patterns in bipolar disorders. J Clin Psychiatry, 67(3), 461467.Google Scholar
Cohen, D (2017). Clozapine and gastrointestinal hypomotility. CNS Drugs, 31(12), 10831091.Google Scholar
Cohen, D, Bogers, JP, van Dijk, D, Bakker, B, Schulte, PF (2012). Beyond white blood cell monitoring: screening in the initial phase of clozapine therapy. J Clin Psychiatry, 73(10), 13071312.Google Scholar
Correll, CU, Rubio, JM, Inczedy-Farkas, G, et al. (2017). Efficacy of 42 pharmacologic cotreatment strategies added to antipsychotic monotherapy in schizophrenia: systematic overview and quality appraisal of the meta-analytic evidence. JAMA Psychiatry, 74, 675684.Google Scholar
Couchman, L, Morgan, EL, Spencer, EP, Flanagan, RJ (2010). Plasma clozapine, norclozapine, and the clozapine:norclozapine ratio in relation to prescribed dose and other factors: data from a therapeutic drug monitoring service, 1993–2007. Ther Drug Monit, 32, 438447.Google Scholar
Crilly, J (2007). The history of clozapine and its emergence in the US market: a review and analysis. Hist Psychiatry, 18(1), 3960.Google Scholar
Davies, LM, Lewis, S, Jones, PB, et al.; CUtLASS team (2007). Cost-effectiveness of first- v. second-generation antipsychotic drugs: results from a randomised controlled trial in schizophrenia responding poorly to previous therapy. Br J Psychiatry, 191, 1422.Google Scholar
Davis, JM, Chen, N, Glick, ID (2003). A meta-analysis of the efficacy of second-generation antipsychotics. Arch Gen Psychiatry, 60(6), 553564.Google Scholar
Demjaha, A, Egerton, A, Murray, RM, et al. (2014). Antipsychotic treatment resistance in schizophrenia associated with elevated glutamate levels but normal dopamine function. Biol Psychiatry, 75(5), e11e13.Google Scholar
Dzahini, O, Singh, N, Taylor, D, Haddad, PM (2018). Antipsychotic drug use and pneumonia: systematic review and meta-analysis. J Psychopharmacol, 32(11), 11671181.Google Scholar
electronic Medicines Compendium (2019). Summary of Product Characteristics: Clorazil. Mylan. Last updated 6 June 2019. Available at: www.medicines.org.uk/emc/product/10290/smpc (last accessed 4.10.19).Google Scholar
Fabrazzo, M, La Pia, S, Monteleone, P, et al. (2002). Is the time course of clozapine response correlated to the time course of clozapine plasma levels? A one-year prospective study in drug-resistant patients with schizophrenia. Neuropsychopharmacology, 27(6), 10501055.Google Scholar
Foley, DL, Mackinnon, A, Morgan, VA, et al. (2015). Effect of age, family history of diabetes, and antipsychotic drug treatment on risk of diabetes in people with psychosis: a population-based cross-sectional study. Lancet Psychiatry, 2(12), 10921098.Google Scholar
Frogley, C, Taylor, D, Dickens, G, Picchioni, M (2011). A systematic review of the evidence of clozapine’s anti-aggressive effects. Int J Neuropsychopharmacol, 15(9), 13511371.Google Scholar
Galling, B, Roldán, A, Hagi, K, et al. (2017). Antipsychotic augmentation vs. monotherapy in schizophrenia: systematic review, meta-analysis and meta-regression analysis.World Psychiatry, 16(1), 7789.Google Scholar
Goldstein, ME, Anderson, VM, Pillai, A, Kydd, RR, Russell, BR (2015). Glutamatergic neurometabolites in clozapine-responsive and -resistant schizophrenia. Int J Neuropsychopharmacol, 18(6), 117.Google Scholar
Gross, G, Wicke, K, Drescher, KU (2013). Dopamine D3 receptor antagonism – still a therapeutic option for the treatment of schizophrenia. Naunyn Schmiedebergs Arch Pharmacol, 386(2), 155166.Google Scholar
Haddad, PM, Correll, CU (2018). The acute efficacy of antipsychotics in schizophrenia: a review of recent meta-analyses. Ther Adv Psychopharmacol, 8(11), 303318.Google Scholar
Haddad, PM, Wieck, A (2004). Antipsychotic-induced hyperprolactinaemia: mechanisms, clinical features and management. Drugs, 64, 22912314.Google Scholar
Hampson, M, Killaspy, H, Mynors-Wallis, L, Meier, R (2011). Outcome measures recommended for use in adult psychiatry. Occasional Paper OP78. London: Royal College of Psychiatrists. Available at: www.rcpsych.ac.uk/files/pdfversion/OP78x.pdf (last accessed 11.8.17).Google Scholar
Healy, D (1997). The Psychopharmcologists: Interviews by David Healy. New York: Chapman and Hall.Google Scholar
Holt, RIG, Peveler, RC (2009). Obesity, serious mental illness and antipsychotic drugs. Diabetes Obes Metabol, 11, 665679.Google Scholar
Howes, OD, Vergunst, F, Gee, S, et al. (2012). Adherence to treatment guidelines in clinical practice: study of antipsychotic treatment prior to clozapine initiation. Br J Psychiatry, 201, 481485.Google Scholar
Howes, OD, McCutcheon, R, Agid, O, et al. (2017). Treatment resistant schizophrenia: Treatment Response and Resistance in Psychosis (TRRIP) working group consensus guidelines on diagnosis and terminology. Am J Psychiatry, 174(3), 216229.Google Scholar
Huang, KL, Fang, CJ, Hsu, CC, et al. (2017). Myocardial infarction risk and antipsychotics use revisited: a meta-analysis of 10 observational studies. J Psychopharmacol, 31(12), 15441555. doi:10.1177/0269881117714047.Google Scholar
Hung, GC, Liu, HC, Yang, SY, et al. (2016). Antipsychotic reexposure and recurrent pneumonia in schizophrenia: a nested case-control study. J Clin Psychiatry, 77, 6066.Google Scholar
Hynes, C, Keating, D, McWilliams, S, et al. (2015). Glasgow Antipsychotic Side-effects Scale for Clozapine – development and validation of a clozapine-specific side-effects scale. Schizophr Res, 168(1–2), 505513.Google Scholar
Ifteni, P, Nielsen, J, Burtea, V, et al. (2014). Effectiveness and safety of rapid clozapine titration in schizophrenia. Acta Psychiatr Scand, 130, 2529.Google Scholar
Ismail, Z, Wessels, AM, Uchida, H, et al. (2012). Age and sex impact clozapine plasma concentrations in inpatients and outpatients with schizophrenia. Am J Geriatr Psychiatry, 20(1), 5360.Google Scholar
Kane, JM, Honigfield, G, Singer, J, Meltzer, HY (1988). Clozapine for the treatment-resistant schizophrenic: a double-blind comparison with chlorpromazine. Arch Gen Psychiatry, 45, 789796.Google Scholar
Kluge, M, Schuld, A, Schacht, A, et al. (2009). Effects of clozapine and olanzapine on cytokine systems are closely linked to weight gain and drug-induced fever. Psychoneuroendocrinology, 34(1), 118128.Google Scholar
Koponen, HJ, Hakko, HH, Saari, KM, et al. (2010). The prevalence and predictive value of individual criteria for metabolic syndrome in schizophrenia: a Northern Finland 1966 Birth Cohort Study. World J Biol Psychiatry, 11(2 Pt 2), 262267.Google Scholar
Krawiecka, M, Goldberg, D, Vaughan, M. (1977). A standardised psychiatric assessment scale for rating chronic psychiatric patients. Acta Psychiatr Scand, 55, 299308.Google Scholar
Kuo, CJ, Yang, SY, Liao, YT, et al. (2013). Second-generation antipsychotic medications and risk of pneumonia in schizophrenia. Schizophr Bull, 39, 648657.Google Scholar
Lahti, RA, Evans, DL, Stratman, NC, Figur, LM (1993). Dopamine D4 versus D2 receptor selectivity of dopamine receptor antagonists: possible therapeutic implications. Eur J Pharmacol, 236(3), 483486.Google Scholar
Lalanne, L, Lutz, PE, Trojak, B, et al. (2016). Medications between psychiatric and addictive disorders. Prog Neuropsychopharmacol Biol Psychiatry, 65, 215223.Google Scholar
Lally, J, Tully, J, Robertson, D, et al. (2016). Augmentation of clozapine with electroconvulsive therapy in treatment resistant schizophrenia: a systematic review and meta-analysis. Schizophr Res, 171(1–3), 215224.Google Scholar
Lambert, M, Haro, JM, Novick, D, et al. (2005). Olanzapine vs. other antipsychotics in actual out-patient settings: six months tolerability results from the European Schizophrenia Out-patient Health Outcomes study. Acta Psychiatr Scand, 111(3), 232243.Google Scholar
Lambiase, PD, de Bono, JP, Schilling, RJ, et al. (2019). British Heart Rhythm Society clinical practice guidelines on the management of patients developing QT prolongation on antipsychotic medication. Arrhythm Electrophysiol Rev, 8(3), 161165.Google Scholar
Laroche, DG, Gaillard, A (2016). Induced obsessive compulsive symptoms (OCS) in schizophrenia patients under atypical 2 antipsychotics (AAPs): review and hypotheses. Psychiatry Res, 246, 119128.Google Scholar
Leucht, S, Cipriani, A, Spijeli, L, et al. (2013). Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. Lancet, 382, 951962.Google Scholar
Lewis, SW, Barnes, TR, Davies, L, et al. (2006). Randomized controlled trial of effect of prescription of clozapine versus other second-generation antipsychotic drugs in resistant schizophrenia. Schizophr Bull, 32(4), 715723.Google Scholar
Li, XB, Tang, YL, Wang, CY, et al. (2015). Clozapine for treatment-resistant bipolar disorder: a systematic review. Bipolar Disord, 17, 235247.Google Scholar
Lieberman, JA, Phillips, M, Gu, H, et al. (2003). Atypical and conventional antipsychotic drugs in treatment-naive first-episode schizophrenia: a 52-week randomized trial of clozapine vs chlorpromazine. Neuropsychopharmacology, 28(5), 9951003.Google Scholar
Lobos, CA, Komossa, K, Rummel-Kluge, C, et al. (2010). Clozapine versus other atypical antipsychotics for schizophrenia. Cochrane Database Syst Rev, (11), CD006633.Google Scholar
Manu, P, Sarpal, D, Muir, O, Kane, JM, Correll, CU (2012). When can patients with potentially life-threatening adverse effects be rechallenged with clozapine? A systematic review of the published literature. Schizophr Res, 134(2–3), 180186.Google Scholar
Manu, P, Lapitskaya, Y, Shaikh, A, Nielsen, J (2018). Clozapine rechallenge after major adverse effects: clinical guidelines based on 259 cases. Am J Ther, 25(2), e218e223.Google Scholar
Mauri, MC, Volonteri, LS, Colasanti, A, et al. (2007). Clinical pharmacokinetics of atypical antipsychotics: a critical review of the relationship between plasma concentrations and clinical response. Clin Pharmacokinet, 46(5), 359388.Google Scholar
McEvoy, JP, Lieberman, JA, Stroup, TS, et al.; CATIE Investigators (2006). Effectiveness of clozapine versus olanzapine, quetiapine, and risperidone in patients with chronic schizophrenia who did not respond to prior atypical antipsychotic treatment. Am J Psychiatry, 163(4), 600610.Google Scholar
Melkersson, K, Lewitt, M, Hall, K (2015). Higher serum concentrations of tyrosine and glutamate in schizophrenia patients treated with clozapine, compared to in those treated with conventional antipsychotics. Neuro Endocrinol Lett, 36(5), 465480.Google Scholar
Meltzer, HY (2013). Update on typical and atypical antipsychotic drugs. Annu Rev Med, 64, 393406.Google Scholar
Meltzer, HY, Alphs, L, Green, AI, et al.; International Suicide Prevention Trial Study Group (2003). Clozapine treatment for suicidality in schizophrenia. Arch Gen Psychiatry, 60, 8291.Google Scholar
Mendoza, MC, Lindenmayer, JP (2009). N-desmethylclozapine: is there evidence for its antipsychotic potential? Clin Neuropharmacol, 32(3), 154157.Google Scholar
Meyer, N, Gee, S, Whiskey, E, et al. (2015). Optimizing outcomes in clozapine rechallenge following neutropenia: a cohort analysis. J Clin Psychiatry, 76(11), e1410e1416.Google Scholar
Miyamoto, S, Miyake, N, Jarskog, LF, Fleischhacker, WW, Lieberman, JA (2012). Pharmacological treatment of schizophrenia: a critical review of the pharmacology and clinical effects of current and future therapeutic agents. Mol Psychiatry, 17(12), 12061227.Google Scholar
Morrison, AP, Pyle, M, Gumley, A, et al. (2019). Cognitive-behavioural therapy for clozapine-resistant schizophrenia: the FOCUS RCT. Health Technol Assess, 23(7), 1144.Google Scholar
Mouchlianitis, E, Bloomfield, MA, Law, V, et al. (2016). Treatment-resistant schizophrenia patients show elevated anterior cingulate cortex glutamate compared to treatment-responsive. Schizophr Bull, 42(3), 744752.Google Scholar
National Institute for Health and Care Excellence (NICE) (2014). Psychosis and schizophrenia in adults: prevention and management. Clinical guideline [CG178]. London: National Institute for Health and Care Excellence. Available at: www.nice.org.uk/guidance/cg178 (last accessed 4.10.19).Google Scholar
Nielsen, J, Meyer, JM (2012). Risk factors for ileus in patients with schizophrenia. Schizophr Bull, 38(3), 592598.Google Scholar
Nielsen, J, Correll, CU, Manu, P, Kane, JM (2013). Termination of clozapine treatment due to medical reasons: when is it warranted and how can it be avoided? J Clin Psychiatry, 74(6), 603613.Google Scholar
Norman, SM, Sullivan, KM, Liu, F, et al. (2017). Blood pressure and heart rate changes during clozapine treatment. Psychiatr Q, 88(3), 545552.Google Scholar
Novick, D, Haro, JM, Perrin, E, Suarez, D, Texeira, JM (2009). Tolerability of outpatient antipsychotic treatment: 36-month results from the European Schizophrenia Outpatient Health Outcomes (SOHO) study. Eur Neuropsychopharmacol, 19(8), 542550.Google Scholar
O’Connor, WT, O’Shea, SD (2015). Clozapine and GABA transmission in schizophrenia disease models: establishing principles to guide treatments. Pharmacol Ther, 150, 4780.Google Scholar
Park, YW, Kim, Y, Lee, JH (2012). Antipsychotic-induced sexual dysfunction and its management. World J Mens Health, 30(3), 153159.Google Scholar
Perdigues, SR, Quecuti, RS, Mane, A, et al. (2016). An observational study of clozapine induced sedation and its pharmacological management. Eur Neuropsychopharmacol, 26(1), 156161.Google Scholar
Pfuhlmann, B, Hiemke, C, Unterecker, S, et al. (2009). Toxic clozapine serum levels during inflammatory reactions. J Clin Psychopharmacol, 29(4), 392394.Google Scholar
Polcwiartek, C, Nielsen, J (2016). The clinical potentials of adjunctive fluvoxamine to clozapine treatment: a systematic review. Psychopharmacology, 233(5), 741750.Google Scholar
Potkin, SG, Basile, VS, Jin, Y, et al. (2003). D1 receptor alleles predict PET metabolic correlates of clinical response to clozapine. Mol Psychiatry, 8, 109113.Google Scholar
Regen, F, Herzog, I, Hahn, E, et al. (2017). Clozapine-induced agranulocytosis: evidence for an immune-mediated mechanism from a patient-specific in-vitro approach. Toxicol Appl Pharmacol, 316, 1016.Google Scholar
Remington, G, Lee, J, Agid, O, et al. (2016). Clozapine’s critical role in treatment resistant schizophrenia: ensuring both safety and use. Expert Opin Drug Saf, 15(9), 11931203.Google Scholar
Roberts, CE, Mortenson, LY, Merrill, DB, et al. (2011). Successful rechallenge with clozapine after eosinophilia. Am J Psychiatry, 168(11), 11471151.Google Scholar
Rogers, DP, Shramko, JK (2000). Therapeutic options in the treatment of clozapine-induced sialorrhea. Pharmacotherapy, 20(9), 10921095.Google Scholar
Ronaldson, KJ, Taylor, AJ, Fitzgerald, PB, et al. (2010). Diagnostic characteristics of clozapine-induced myocarditis identified by an analysis of 38 cases and 47 controls. J Clin Psychiatry, 71(8), 976981.Google Scholar
Ronaldson, KJ, Fitzgerald, PB, Taylor, AJ, et al. (2012). Rapid clozapine dose titration and concomitant sodium valproate increase the risk of myocarditis with clozapine: a case-control study. Schizophr Res, 141(2–3), 173178.Google Scholar
Rostami-Hodjegan, A, Amin, AM, Spencer, EP, et al. (2004). Influence of dose, cigarette smoking, age, sex, and metabolic activity on plasma clozapine concentrations: a predictive model and nomograms to aid clozapine dose adjustment and to assess compliance in individual patients. J Clin Psychopharmacol, 24(1), 7078.Google Scholar
Roth, BL, Driscol, J (2013). PDSP Ki Database. Psychoactive Drug Screening Program (PDSP). University of North Carolina at Chapel Hill and the United States National Institute of Mental Health. Retrieved 10.10.13 from ‘Archived copy’. Archived from the original on 8.11.13. Retrieved 25.11.13.Google Scholar
Samara, MT, Dold, M, Gianatsi, M, et al. (2016). Efficacy, acceptability, and tolerability of antipsychotics in treatment-resistant schizophrenia: a network meta-analysis. JAMA Psychiatry, 73(3), 199210.Google Scholar
Seeman, P (2014). Clozapine, a fast-off-D2 antipsychotic. ACS Chem Neurosci, 5, 2429.Google Scholar
Selent, J, Lopez, L, Sanz, F, Pastor, M (2008). Multi-receptor binding profile of clozapine and olanzapine: a structural study based on the new β2 adrenergic receptor template. ChemMedChem, 3, 11941198.Google Scholar
Shirazi, A, Stubbs, B, Gomez, L, et al. (2016). Prevalence and predictors of clozapine-associated constipation: a systematic review and meta-analysis. Int J Mol Sci, 17(6), 863.Google Scholar
Siskind, D, McCartney, L, Goldschlager, R, Kisely, S (2016). Clozapine v. first- and second-generation antipsychotics in treatment refractory schizophrenia: systematic review and meta-analysis. Br J Psychiatry, 209(5), 385392.Google Scholar
Siskind, D, Siskind, V, Kisely, S (2017). Clozapine response rates among people with treatment-resistant schizophrenia: data from a systematic review and meta-analysis. Can J Psychiatry, 62(11), 772777.Google Scholar
Sommer, IE, Begemann, MJ, Temmerman, A, Leucht, S (2012). Pharmacological augmentation strategies for schizophrenia patients with insufficient response to clozapine: a quantitative literature review. Schizophr Bull, 38(5), 10031011.Google Scholar
Sommer, IE, van Westrhenen, R, Begemann, MJH, et al. (2014). Efficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia: an update. Schizophr Bull, 40(1), 181191.Google Scholar
Stoecker, ZR, George, WT, O’Brien, JB, et al. (2017). Clozapine usage increases the incidence of pneumonia compared with risperidone and the general population: a retrospective comparison of clozapine, risperidone, and the general population in a single hospital over 25 months. Int Clin Psychopharmacol, 32(3), 155160.Google Scholar
Suppes, T, Webb, A, Paul, B, et al. (1999). Clinical outcome in a randomized 1-year trial of clozapine versus treatment as usual for patients with treatment-resistant illness and a history of mania. Am J Psychiatry, 156, 11641169.Google Scholar
Suzuki, T, Remington, G, Uchida, H, et al. (2011a). Mangement of schizophrenia in late life with antipsychotic medications. A qualitative review. Drugs Aging, 28(12), 961980.Google Scholar
Suzuki, T, Uchida, H, Watanabe, K, Kashima, H (2011b). Factors associated with response to clozapine in schizophrenia: a review. Psychopharmacol Bull, 44(1), 3260.Google Scholar
Tanahashi, S, Yamamura, S, Nakagawa, M, Motomura, E, Okada, M (2012). Clozapine, but not haloperidol, enhances glial D-serine and L-glutamate release in rat frontal cortex and primary cultured astrocytes. Br J Pharmacol, 165(5), 15431555.Google Scholar
Testani, M Jr (1994). Clozapine-induced orthostatic hypotension treated with fludrocortisone. J Clin Psychiatry, 55(11), 497498.Google Scholar
Tiihonen, J, Lönnqvist, J, Wahlbeck, K, et al. (2009). 11-year follow-up of mortality in patients with schizophrenia: a population-based cohort study (FIN11 study). Lancet, 374(9690), 620627.Google Scholar
Tiihonen, J, Haukka, J, Taylor, M, et al. (2011). A nationwide cohort study of oral and depot antipsychotics after first hospitalization for schizophrenia. Am J Psychiatry, 168(6), 603609.Google Scholar
Tiihonen, J, Mittendorfer-Rutz, E, Majak, M, et al. (2017). Real-world effectiveness of antipsychotic treatments in a nationwide cohort of 29823 patients with schizophrenia. JAMA Psychiatry, 74(7), 686693.Google Scholar
Veerman, SR, Schulte, PF, Begemann, MJ, de Haan, L (2014). Non-glutamatergic clozapine augmentation strategies: a review and meta-analysis. Pharmacopsychiatry, 47(7), 231–8.Google Scholar
World Health Organization (WHO) (2017). WHO Model Lists of Essential Medicines, 20th list, 24.1. Geneva: World Health Organization. Available at: www.who.int/medicines/publications/essentialmedicines/en/ (last accessed 9.8.17).Google Scholar
Wu, CS, Wang, SC, Gau, SSF, Tsai, HJ, Cheng, YC (2013). Association of stroke with the receptor-binding profiles of antipsychotics – a case-crossover study. Biol Psychiatry, 73, 414421.Google Scholar
Wu, CS, Tsai, YT, Tsai, HJ (2015). Antipsychotic drugs and the risk of ventricular arrhythmia and/or sudden cardiac death: a nation-wide case-crossover study. J Am Heart Assoc, 4(2), e001568.Google Scholar
Wu, Y, Blichowski, M, Daskalakis, ZJ, et al. (2011). Evidence that clozapine directly interacts on the GABAB receptor. Neuroreport, 22(13), 637641.Google Scholar
Xiang, YQ, Zhang, ZJ, Weng, YZ, et al. (2006). Serum concentrations of clozapine and norclozapine in the prediction of relapse of patients with schizophrenia. Schizophr Res, 83(2–3), 201210.Google Scholar
Yang, SY, Liao, YT, Liu, HC, et al. (2013). Antipsychotic drugs, mood stabilizers, and risk of pneumonia in bipolar disorder: a nationwide case-control study. J Clin Psychiatry, 74, e79e86.Google Scholar
Yoshimura, B, Yada, Y, So, R, Takaki, M, Yamada, N (2017). The critical treatment window of clozapine in treatment-resistant schizophrenia: secondary analysis of an observational study. Psychiatry Res, 250, 6570.Google Scholar
Yusufi, B, Mukherjee, S, Flanagan, R, et al. (2007). Prevalence and nature of side effects during clozapine maintenance treatment and the relationship with clozapine dose and plasma concentration. Int J Psychopharmacol, 22, 238243.Google Scholar
Zheng, W, Xiang, YT, Yang, XH, Xiang, YQ, de Leon, J (2017). Clozapine augmentation with antiepileptic drugs for treatment-resistant schizophrenia: a meta-analysis of randomized controlled trials. J Clin Psychiatry, 78(5), e498e505.Google Scholar

References

Abou-Saleh, MT, Müller-Oerlinghausen, B, Coppen, AJ (2017). Lithium in the episode and suicide prophylaxis and in augmenting strategies in patients with unipolar depression. Int J Bipolar Disord, 5, 11.Google Scholar
Ahrens, B, Müller-Oerlinghausen, B (2001). Does lithium exert an independent antisuicidal effect? Pharmacopsychiatry, 34(4), 132136.Google Scholar
Aubrey, RE, Scott, L, Cassidy, E (2017). Lithium monitoring patterns in the United Kingdom and Ireland: can shared care agreements play a role in monitoring quality? A systematic review. Ir J Psychol Med, 34, 127140.Google Scholar
Baird-Gunning, J, Lea-Henry, T, Hoegberg, LCG, Gosselin, S, Roberts, DM (2017). Lithium poisoning. J Intensive Care Med, 32, 249263.Google Scholar
Barker, WA, Scott, J, Eccleston, D (1987). The Newcastle chronic depression study: results of a treatment regime. Int Clin Psychopharmacol, 2(3), 261272.Google Scholar
Bauer, M, Gitlin, M (2016). The Essential Guide to Lithium Treatment. Berlin: Springer International Publishing AG.Google Scholar
Bauer, M, Dell’osso, L, Kasper, S, et al. (2013). Extended-release quetiapine fumarate (quetiapine XR) monotherapy and quetiapine XR or lithium as add-on to antidepressants in patients with treatment-resistant major depressive disorder. J Affect Disord, 151, 209219.Google Scholar
Bell, AJ, Cole, A, Eccleston, D, Ferrier, IN (1993). Lithium neurotoxicity at normal therapeutic levels. Br J Psychiatry, 162, 689692.Google Scholar
Beurel, E, Jope, RS (2006). The paradoxical pro- and anti-apoptotic actions of GSK3 in the intrinsic and extrinsic apoptosis signaling pathways. Prog Neurobiol, 79(4), 173189.Google Scholar
Beurel, E, Grieco, SF, Jope, RS (2015). Glycogen synthase kinase-3 (GSK3): regulation, actions, and diseases. Pharmacol Ther, 148, 114131.Google Scholar
BNF (2017). British National Formulary. London: NICE.Google Scholar
Brunello, N, Tascedda, F (2003). Cellular mechanisms and second messengers: relevance to the psychopharmacology of bipolar disorders. Int J Neuropsychopharmacol, 6, 181189.Google Scholar
Cade, JF (1949). Lithium salts in the treatment of psychotic excitement. Med J Aust, 2, 349352.Google Scholar
Can, A, Schulze, TG, Gould, TD (2014). Molecular actions and clinical pharmacogenetics of lithium therapy. Pharmacol Biochem Behav, 123, 316.Google Scholar
Cipriani, A, Smith, K, Burgess, S, et al. (2006). Lithium versus antidepressants in the long-term treatment of unipolar affective disorder. Cochrane Database Syst Rev, (4), CD003492.Google Scholar
Cipriani, A, Barbui, C, Salanti, G, et al. (2011). Comparative efficacy and acceptability of antimanic drugs in acute mania: a multiple-treatments meta-analysis. Lancet, 378, 13061315.Google Scholar
Cipriani, A, Hawton, K, Stockton, S, Geddes, JR (2013). Lithium in the prevention of suicide in mood disorders: updated systematic review and meta-analysis. BMJ, 346, f3646.Google Scholar
Cleare, A, Pariante, CM, Young, AH, et al. (2015). Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 2008 British Association for Psychopharmacology guidelines. J Psychopharmacol, 29, 459525.Google Scholar
Cogen, PH, Whybrow, PC (2006). Lithium: a fascinating element in neuropsychiatry. In Bauer, M, Grof, P, Muller-Oerlinghausen, B, eds., Lithium in Psychiatry: The Comprehensive Guide. London: Informa Healthcare.Google Scholar
Ferrie, L, Young, AH, McQuade, R (2005). Effect of chronic lithium and withdrawal from chronic lithium on presynaptic dopamine function in the rat. J Psychopharmacol, 19(3), 229234.Google Scholar
Franks, M, Macritchie, KA, Mahmood, T, Young, AH (2008). Bouncing back: is the bipolar rebound phenomenon peculiar to lithium? A retrospective naturalistic study. J Psychopharmacol, 22(4), 452456. doi:10.1177/0269881107085238.Google Scholar
Gershon, S, Chengappa, KN, Malhi, GS (2009). Lithium specificity in bipolar illness: a classic agent for the classic disorder. Bipolar Disord, 11(Suppl 2), 3444.Google Scholar
Ghasemi, M, Dehpour, AR (2011). The NMDA receptor/nitric oxide pathway: a target for the therapeutic and toxic effects of lithium. Trends Pharmacol Sci, 32, 420434.Google Scholar
Goodwin, GM (1994). Recurrence of mania after lithium withdrawal. Implications for the use of lithium in the treatment of bipolar affective disorder. Br J Psychiatry, 164(2), 149152.Google Scholar
Goodwin, GM, Haddad, PM, Ferrier, IN, et al. (2016). Evidence-based guidelines for treating bipolar disorder: revised third edition recommendations from the British Association for Psychopharmacology. J Psychopharmacol, 30, 495553.Google Scholar
Grandjean, EM, Aubry, JM (2009). Lithium: updated human knowledge using an evidence-based approach. Part II: Clinical pharmacology and therapeutic monitoring. CNS Drugs, 23, 331349.Google Scholar
Hajek, T, Cullis, J, Novak, T, et al. (2012). Hippocampal volumes in bipolar disorders: opposing effects of illness burden and lithium treatment. Bipolar Disord, 14, 261270.Google Scholar
Hajek, T, Bauer, M, Simhandl, C, et al. (2014). Neuroprotective effect of lithium on hippocampal volumes in bipolar disorder independent of long-term treatment response. Psychol Med, 44, 507517.Google Scholar
Hanson, ND, Nemeroff, CB, Owens, MJ (2011). Lithium, but not fluoxetine or the corticotropin-releasing factor receptor 1 receptor antagonist R121919, increases cell proliferation in the adult dentate gyrus. J Pharmacol Exp Ther, 337, 180186.Google Scholar
Hayes, JF, Marston, L, Walters, K, et al. (2016a). Lithium vs. valproate vs. olanzapine vs. quetiapine as maintenance monotherapy for bipolar disorder: a population-based UK cohort study using electronic health records. World Psychiatry, 15, 5358.Google Scholar
Hayes, JF, Pitman, A, Marston, L, et al. (2016b). Self-harm, unintentional injury, and suicide in bipolar disorder during maintenance mood stabilizer treatment: a UK population-based electronic health records study. JAMA Psychiatry, 73(6), 630637. doi:10.1001/jamapsychiatry.2016.0432.Google Scholar
Jope, RS, Nemeroff, CB (2016). Lithium to the rescue. Cerebrum, 2016, cer-02-16.Google Scholar
Kessing, LV, Sondergard, L, Forman, JL, Andersen, PK (2008). Lithium treatment and risk of dementia. Arch Gen Psychiatry, 65, 13311335.Google Scholar
Kessing, LV, Forman, JL, Andersen, PK (2010). Does lithium protect against dementia? Bipolar Disord, 12, 8794.Google Scholar
Kessing, LV, Vradi, E, Andersen, PK (2014). Starting lithium prophylaxis early v. late in bipolar disorder. Br J Psychiatry, 205(3), 214220. doi:10.1192/bjp.bp.113.142802.Google Scholar
Machado-Vieira, R, Gattaz, WF, Zanetti, MV, et al. (2015). A longitudinal (6-week) 3T (1)H-MRS study on the effects of lithium treatment on anterior cingulate cortex metabolites in bipolar depression. Eur Neuropsychopharmacol, 25, 23112317.Google Scholar
Malhi, GS, Outhred, T (2016). Therapeutic mechanisms of lithium in bipolar disorder: recent advances and current understanding. CNS Drugs, 30, 931949.Google Scholar
Malhi, GS, Adams, D, Berk, M (2009). Is lithium in a class of its own? A brief profile of its clinical use. Aust N Z J Psychiatry, 43, 10961104.Google Scholar
Malhi, GS, Tanious, M, Das, P, Coulston, CM, Berk, M (2013). Potential mechanisms of action of lithium in bipolar disorder. Current understanding. CNS Drugs, 27(2), 135153. doi:10.1007/s40263-013-0039-0.Google Scholar
Malhi, GS, Gessler, D, Outhred, T (2017). The use of lithium for the treatment of bipolar disorder: recommendations from clinical practice guidelines. J Affect Disord, 217, 266280.Google Scholar
Mander, AJ, Loudon, JB (1988). Rapid recurrence of mania following abrupt discontinuation of lithium. Lancet, 2(8601), 1517.Google Scholar
National Institute for Health and Care Excellence (NICE) (2014). Bipolar disorder: assessment and management. Clinical Guidelines [CG185]. London: National Institute for Health and Care Excellence.Google Scholar
Nolen, WA, Licht, RW, Young, AH, et al.; Task Force on the treatment with lithium (2019). What is the optimal serum level for lithium in the maintenance treatment of bipolar disorder? A systematic review and recommendations from the ISBD/IGSLI Task Force on treatment with lithium. Bipolar Disord, 21(5), 394409. doi:10.1111/bdi.12805.Google Scholar
Severus, E, Bauer, M, Geddes, J (2018). Efficacy and effectiveness of lithium in the long-term treatment of bipolar disorders: an update 2018. Pharmacopsychiatry, 51(5), 173176. doi:10.1055/a-0627-7489.Google Scholar
Shelley, R (2004). Affective disorders: 2. Lithium and anticonvulsants. In King, DJ, ed., Seminars in Psychopharmacology, 2nd ed. London: Royal College of Psychiatrists.Google Scholar
Song, J, Sjölander, A, Joas, E, et al. (1917). Suicidal behavior during lithium and valproate treatment: a within-individual 8-year prospective study of 50,000 patients with bipolar disorder. Am J Psychiatry, 174(8), 795802. doi:10.1176/appi.ajp.2017.16050542.Google Scholar
Taylor, D, Paton, C, Kapur, S (2015). The Maudsley Prescribing Guidelines in Psychiatry, 12th ed. Hoboken: Wiley Blackwell.Google Scholar
Tiihonen, J, Tanskanen, A, Hoti, F, et al. (2017). Pharmacological treatments and risk of readmission to hospital for unipolar depression in Finland: a nationwide cohort study. Lancet Psychiatry, 4(7), 547553. doi:10.1016/S2215-0366(17)30134-7.Google Scholar
Viguera, AC, Nonacs, R, Cohen, LS, et al. (2000). Risk of recurrence of bipolar disorder in pregnant and nonpregnant women after discontinuing lithium maintenance. Am J Psychiatry, 157, 179184.Google Scholar
Wakita, M, Nagami, H, Takase, Y, et al. (2015). Modifications of excitatory and inhibitory transmission in rat hippocampal pyramidal neurons by acute lithium treatment. Brain Res Bull, 117, 3944.Google Scholar
Weeks, ME, Larson, ME (1937). J.A. Arfwedson and his services to chemistry. J Chem Educ, 14, 403407.Google Scholar
Weisler, RH, Nolen, WA, Neijber, A, Hellqvist, A, Paulsson, B; Trial 144 Study Investigators (2011). Continuation of quetiapine versus switching to placebo or lithium for maintenance treatment of bipolar I disorder (Trial 144: a randomized controlled study). J Clin Psychiatry, 72(11), 14521464. doi:10.4088/JCP.11m06878. Erratum in: J Clin Psychiatry 2014, 75(3), 290.Google Scholar
World Health Organization (2000). Preventing Suicide: A Resource for General Physicians. Available at: www.who.int/mental_health/media/en/56.pdf (last accessed 22.8.19).Google Scholar
Young, AH (2014). Lithium and suicide. Lancet Psychiatry, 1(6), 483484. doi:10.1016/S2215-0366(14)70320-7.Google Scholar

References

Acharya, S, Bussel, JB (2000). Hematologic toxicity of sodium valproate. J Pediatr Hematol Oncol, 22(1), 6265.Google Scholar
Aiken, CB, Orr, C (2010). Rechallenge with lamotrigine after a rash: a prospective case series and review of the literature. Psychiatry, 7, 2732.Google Scholar
Aliyev, NA, Aliyev, ZN (2008). Valproate (depakine-chrono) in the acute treatment of outpatients with generalized anxiety disorder without psychiatric comorbidity: randomized, double-blind placebo-controlled study. Eur Psychiatry, 23, 109114.Google Scholar
Allen, MH, Hirschfeld, RM, Wozniak, PJ, Baker, JD, Bowden, CL (2006). Linear relationship of valproate serum concentration to response and optimal serum levels for acute mania. Am J Psychiatry, 163, 272275.Google Scholar
Angus-Lepan, H, Liu, R (2018). Weighing the risks of valproate in women who could become pregnant. BMJ, 361, k1596.Google Scholar
Azorin, JM, Bowden, CL, Garay, RP, et al. (2010). Possible new ways in the pharmacological treatment of bipolar disorder and comorbid alcoholism. Neuropsychiatr Dis Treat, 6, 3746.Google Scholar
Ballenger, JC, Post, RM (1978). Therapeutic effects of carbamazepine in affective illness: a preliminary report. Commun Psychopharmacol, 2, 159175.Google Scholar
Ballenger, JC, Post, RM (1980). Carbamazepine in manic-depressive illness: a new treatment. Am J Psychiatry, 137, 782–790.Google Scholar
Belcastro, V, D’Egidio, C, Striano, P, Verrotti, A (2013). Metabolic and endocrine effects of valproic acid chronic treatment. Epilepsy Res, 107, 18.Google Scholar
Bjerkedal, T, Czeizel, A, Goujard, J, et al. (1982). Valproic acid and spina bifida. Lancet, 2, 1096.Google Scholar
BNF (2019). British National Formulary (online). London: BMJ Group and Pharmaceutical Press. Available at: www.medicinescomplete.com (last accessed 13.8.19).Google Scholar
Bond, DJ, Lam, RW, Yatham, LN (2010). Divalproex sodium versus placebo in the treatment of acute bipolar depression: a systematic review and meta-analysis. J Affect Disord, 124, 228234.Google Scholar
Bowden, CL, Asnis, GM, Ginsberg, LD, et al. (2004). Safety and tolerability of lamotrigine for bipolar disorder. Drug Saf, 27, 173184.Google Scholar
Brodie, MJ (1992). Lamotrigine. Lancet, 339, 13971400.Google Scholar
Brodie, MJ (2010). Antiepileptic drug therapy the story so far. Seizure, 19, 650655.Google Scholar
Brodie, MJ, Mintzer, S, Pack, AM, et al. (2013). Enzyme induction with antiepileptic drugs: cause for concern? Epilepsia, 54, 1127.Google Scholar
Bromley, R, Weston, J, Adab, N, et al. (2014). Treatment for epilepsy in pregnancy: neurodevelopmental outcomes in the child. Cochrane Database Syst Rev, (10), CD010236.Google Scholar
Campbell, E, Kennedy, F, Russell, A, et al. (2014). Malformation risks of antiepileptic drug monotherapies in pregnancy: updated results from the UK and Ireland Epilepsy and Pregnancy Registers. J Neurol Neurosurg Psychiatry, 85, 10291034.Google Scholar
Carrigan, PJ, Brinker, DR, Cavanaugh, JH, Lamm, JE, Cloyd, JC (1990). Absorption characteristics of a new valproate formulation: divalproex sodium-coated particles in capsules (Depakote Sprinkle). J Clin Pharmacol, 30, 743747.Google Scholar
Chen, PS, Peng, GS, Li, G, et al. (2006). Valproate protects dopaminergic neurons in midbrain neuron/glia cultures by stimulating the release of neurotrophic factors from astrocytes. Mol Psychiatry, 11, 11161125.Google Scholar
Christensen, J, Gronborg, TK, Sorensen, MJ, et al. (2013). Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA, 309, 16961703.Google Scholar
Chung, S, Wang, N, Hank, N (2007). Comparative retention rates and long-term tolerability of new antiepileptic drugs. Seizure, 16, 296304.Google Scholar
Cipriani, A, Barbui, C, Salanti, G, et al. (2011). Comparative efficacy and acceptability of antimanic drugs in acute mania: a multiple-treatments meta-analysis. Lancet, 378, 13061315.Google Scholar
Citrome, L, Volavka, J (2011). Pharmacological management of acute and persistent aggression in forensic psychiatry settings. CNS Drugs, 25, 10091021.Google Scholar
Cleare, A, Pariante, CM, Young, AH, et al.; Members of the Consensus Meeting (2015). Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 2008 British Association for Psychopharmacology guidelines. J Psychopharmacol, 29, 459525.Google Scholar
Cohen, AF, Land, GS, Breimer, DD, et al. (1987). Lamotrigine, a new anticonvulsant: pharmacokinetics in normal humans. Clin Pharmacol Ther, 42, 535541.Google Scholar
Cohen, MJ, Meador, KJ, Browning, N, et al. (2011). Fetal antiepileptic drug exposure: motor, adaptive, and emotional/behavioral functioning at age 3 years. Epilepsy Behav, 22, 240246.Google Scholar
Cohen, MJ, Meador, KJ, Browning, N, et al.; NEAD study group (2013). Fetal antiepileptic drug exposure: adaptive and emotional/behavioral functioning at age 6 years. Epilepsy Behav, 29, 308315.Google Scholar
Correll, CU, Rubio, JM, Inczedy-Farkas, G, et al. (2017). Efficacy of 42 pharmacologic cotreatment strategies added to antipsychotic monotherapy in schizophrenia: systematic overview and quality appraisal of the meta-analytic evidence. JAMA Psychiatry, 74, 675684.Google Scholar
Crawford, MJ, Sanatinia, R, Barrett, B, et al. (2018). Lamotrigine for people with borderline personality disorder: a RCT. Health Technol Assess, 22, 168.Google Scholar
Cummings, C, Stewart, M, Stevenson, M, Morrow, J, Nelson, J (2011). Neurodevelopment of children exposed in utero to lamotrigine, sodium valproate and carbamazepine. Arch Dis Child, 96, 643647.Google Scholar
Davis, LL, Ryan, W, Adinoff, B, Petty, F (2000). Comprehensive review of the psychiatric uses of valproate. J Clin Psychopharmacol, 20, 1S17S.Google Scholar
Dias, VV, Balanza-Martinez, V, Soeiro-de-Souza, MG, et al. (2012). Pharmacological approaches in bipolar disorders and the impact on cognition: a critical overview. Acta Psychiatr Scand, 126, 315331.Google Scholar
Drevets, WC (2000). Neuroimaging studies of mood disorders. Biol Psychiatry, 48, 813829.Google Scholar
Drugbank (2019a). Lamotrigine. Available at: www.drugbank.ca/drugs/DB00555 (last accessed 6.8.19).Google Scholar
Drugbank (2019b). Carbamazepine. Available at: www.drugbank.ca/drugs/DB00564 (last accessed 6.8.19).Google Scholar
electronic Medicines Compendium (2019a). Summary of Product Characteristics: Depakote. SANOFI. Available at: www.medicines.org.uk/emc/product/6113/smpc (last accessed 6.8.19).Google Scholar
electronic Medicines Compendium (2019b). Summary of Product Characteristics: Lamotrigine. Accord Healthcare Limited. Available at: www.medicines.org.uk/emc/product/6091/smpc (last accessed 6.8.19).Google Scholar
electronic Medicines Compendium (2019c). Summary of Product Characteristics: Carbamazepine. Novartis Pharmaceuticals UK Ltd. Available at: www.medicines.org.uk/emc/product/1040/smpc (last accessed 6.8.19).Google Scholar
Elkjaer, LS, Bech, BH, Sun, Y, Laursen, TM, Christensen, J (2018). Association between prenatal valproate exposure and performance on standardized language and mathematics tests in school-aged children. JAMA Neurol, 19, 19.Google Scholar
European Medicines Agency (2018). New measures to avoid valproate exposure in pregnancy endorsed. Press release, 23 March 2018. Available at: www.ema.europa.eu/docs/en_GB/document_library/Press_release/2018/03/WC500246391.pdf (last accessed 22.8.19).Google Scholar
Fleming, J, Chetty, M (2005). Psychotropic drug interactions with valproate. Clin Neuropharmacol, 28, 96101.Google Scholar
Franks, M, Macritchie, KA, Mahmood, T, Young, AH (2008). Bouncing back: is the bipolar rebound phenomenon peculiar to lithium? A retrospective naturalistic study. J Psychopharmacol, 22, 452456.Google Scholar
Frye, MA, Ketter, TA, Kimbrell, TA, et al. (2000). A placebo-controlled study of lamotrigine and gabapentin monotherapy in refractory mood disorders. J Clin Psychopharmacol, 20, 607614.Google Scholar
Geddes, JR, Calabrese, JR, Goodwin, GM (2009). Lamotrigine for treatment of bipolar depression: independent meta-analysis and meta-regression of individual patient data from five randomised trials. Br J Psychiatry, 194, 49.Google Scholar
Geddes, JR, Gardiner, A, Rendell, J, et al.; CEQUEL Investigators and Collaborators (2016). Comparative evaluation of quetiapine plus lamotrigine combination versus quetiapine monotherapy (and folic acid versus placebo) in bipolar depression (CEQUEL): a 2 × 2 factorial randomised trial. Lancet Psychiatry, 3, 3139.Google Scholar
Ghabrash, MF, Comai, S, Tabaka, J, et al. (2016). Valproate augmentation in a subgroup of patients with treatment-resistant unipolar depression. World J Biol Psychiatry, 17, 165170.Google Scholar
Ghodke-Puranik, Y, Thorn, CF, Lamba, JK, et al. (2013). Valproic acid pathway: pharmacokinetics and pharmacodynamics. Pharmacogenet Genomics, 23, 236241.Google Scholar
Goodwin, GM, Bowden, CL, Calabrese, JR, et al. (2004). A pooled analysis of 2 placebo-controlled 18-month trials of lamotrigine and lithium maintenance in bipolar I disorder. J Clin Psychiatry, 65, 432441.Google Scholar
Goodwin, GM, Haddad, PM, Ferrier, IN, et al. (2016). Evidence-based guidelines for treating bipolar disorder: revised third edition recommendations from the British Association for Psychopharmacology. J Psychopharmacol, 30, 495553.Google Scholar
Grunze, H, Kotlik, E, Costa, R, et al. (2015). Assessment of the efficacy and safety of eslicarbazepine acetate in acute mania and prevention of recurrence: experience from multicentre, double-blind, randomised phase II clinical studies in patients with bipolar disorder I. J Affect Disord, 174, 7082.Google Scholar
Hammond, CJ, Niciu, MJ, Drew, S, Arias, AJ (2015). Anticonvulsants for the treatment of alcohol withdrawal syndrome and alcohol use disorders. CNS Drugs, 29, 293311.Google Scholar
Haymond, J, Ensom, MH (2010). Does valproic acid warrant therapeutic drug monitoring in bipolar affective disorder? Ther Drug Monit, 32, 1929.Google Scholar
Hernandez-Diaz, S, Smith, CR, Shen, A, et al.; North American AED Pregnancy Registry (2012). Comparative safety of antiepileptic drugs during pregnancy. Neurology, 78, 16921699.Google Scholar
Huband, N, Ferriter, M, Nathan, R, Jones, H (2010). Antiepileptics for aggression and associated impulsivity. Cochrane Database Syst Rev, (2), CD003499.Google Scholar
Hurley, SC (2002). Lamotrigine update and its use in mood disorders. Ann Pharmacother, 36, 860873.Google Scholar
Jentink, J, Loane, MA, Dolk, H, et al.; EUROCAT Antiepileptic Study Working Group (2010). Valproic acid monotherapy in pregnancy and major congenital malformations. N Engl J Med, 362, 21852193.Google Scholar
Kaye, NS, Graham, J, Roberts, J, Thompson, T, Nanry, K (2007). Effect of open-label lamotrigine as monotherapy and adjunctive therapy on the self-assessed cognitive function scores of patients with bipolar I disorder. J Clin Psychopharmacol, 27, 387391.Google Scholar
Ketter, TA, Post, RM, Parekh, PI, Worthington, K (1995). Addition of monoamine oxidase inhibitors to carbamazepine: preliminary evidence of safety and antidepressant efficacy in treatment-resistant depression. J Clin Psychiatry, 56(10), 471475.Google Scholar
Kleindienst, N, Greil, W (2000). Differential efficacy of lithium and carbamazepine in the prophylaxis of bipolar disorder: results of the MAP study. Neuropsychobiology, 42(Suppl 1), 210.Google Scholar
Klotz, U, Antonin, KH (1977). Pharmacokinetics and bioavailability of sodium valproate. Clin Pharmacol Ther, 21, 736743.Google Scholar
Kramlinger, KG, Post, RM (1989). The addition of lithium to carbamazepine. Antidepressant efficacy in treatment-resistant depression. Arch Gen Psychiatry, 46, 794800.Google Scholar
Leach, MJ, Marden, CM, Miller, AA (1986). Pharmacological studies on lamotrigine, a novel potential antiepileptic drug: II. Neurochemical studies on the mechanism of action. Epilepsia, 27, 490497.Google Scholar
Lempérière, T (2001). Brief history of the development of valproate in bipolar disorders. Encephale, 27, 365372.Google Scholar
Leucht, S, Helfer, B, Dold, M, Kissling, W, McGrath, J (2014). Carbamazepine for schizophrenia. Cochrane Database Syst Rev, (5), CD001258.Google Scholar
Lieb, K, Vollm, B, Rucker, G, Timmer, A, Stoffers, JM (2010). Pharmacotherapy for borderline personality disorder: Cochrane systematic review of randomised trials. Br J Psychiatry, 196, 412.Google Scholar
Lloyd, KA (2013). A scientific review: mechanisms of valproate-mediated teratogenesis. Biosci Horizons, 6, hzt003-hzt03.Google Scholar
Locharernkul, C, Shotelersuk, V, Hirankarn, N (2011). Pharmacogenetic screening of carbamazepine-induced severe cutaneous allergic reactions. J Clin Neurosci, 18, 12891294.Google Scholar
Löscher, W (1999). The discovery of valproate. In Löscher, W, ed., Valproate. Basel: Birkhäuser Basel, pp. 13.Google Scholar
Meador, K, Reynolds, MW, Crean, S, Fahrbach, K, Probst, C (2008). Pregnancy outcomes in women with epilepsy: a systematic review and meta-analysis of published pregnancy registries and cohorts. Epilepsy Res, 81, 113.Google Scholar
Molgaard-Nielsen, D, Hviid, A (2011). Newer-generation antiepileptic drugs and the risk of major birth defects. JAMA, 305, 19962002.Google Scholar
Mula, M, Pini, S, Cassano, GB (2007). The role of anticonvulsant drugs in anxiety disorders: a critical review of the evidence. J Clin Psychopharmacol, 27, 263–272.Google Scholar
National Institute for Health and Clinical Excellence (NICE) (2009). Borderline personality disorder: recognition and management. Clinical guideline [CG78]. London: National Institute for Health and Care Excellence. Available at: www.nice.org.uk/guidance/cg78 (last accessed 22.8.19).Google Scholar
Nishino, S, Ohtomo, K, Numata, Y, et al. (2012). Divergent effects of lithium and sodium valproate on brain-derived neurotrophic factor (BDNF) production in human astrocytoma cells at therapeutic concentrations. Prog Neuropsychopharmacol Biol Psychiatry, 39, 1722.Google Scholar
Nugent, AC, Carlson, PJ, Bain, EE, et al. (2013). Mood stabilizer treatment increases serotonin type 1 A receptor binding in bipolar depression. J Psychopharmacol, 27, 894902.Google Scholar
Okuma, T, Kishimoto, A (1998). A history of investigation on the mood stabilizing effect of carbamazepine in Japan. Psychiatry Clin Neurosci, 52, 312.Google Scholar
Okuma, T, Kishimoto, A, Inoue, K, Matsumoto, H, Ogura, A (1973). Anti-manic and prophylactic effects of carbamazepine (Tegretol) on manic depressive psychosis. A preliminary report. Folia Psychiatr Neurol Jpn, 27, 283297.Google Scholar
Ornoy, A (2009). Valproic acid in pregnancy: how much are we endangering the embryo and fetus? Reprod Toxicol, 28, 110.Google Scholar
Perugi, G, Toni, C, Frare, F, et al. (2002). Effectiveness of adjunctive gabapentin in resistant bipolar disorder: is it due to anxious-alcohol abuse comorbidity? J Clin Psychopharmacol, 22, 584591.Google Scholar
Peterson, GM, Naunton, M (2005). Valproate: a simple chemical with so much to offer. J Clin Pharm Ther, 30, 417421.Google Scholar
Rajkowska, G (2000). Postmortem studies in mood disorders indicate altered numbers of neurons and glial cells. Biol Psychiatry, 48, 766777.Google Scholar
Ramsay, RE, Pellock, JM, Garnett, WR, et al. (1991). Pharmacokinetics and safety of lamotrigine (Lamictal) in patients with epilepsy. Epilepsy Res, 10, 191200.Google Scholar
Rapoport, SI, Basselin, M, Kim, HW, Rao, JS (2009). Bipolar disorder and mechanisms of action of mood stabilizers. Brain Res Rev, 61, 185209.Google Scholar
Reid, JG, Gitlin, MJ, Altshuler, LL (2013). Lamotrigine in psychiatric disorders. J Clin Psychiatry, 74, 675684.Google Scholar
Robert, E, Guibaud, P (1982). Maternal valproic acid and congenital neural tube defects. Lancet, 2, 937.Google Scholar
Rosenberg, G (2007). The mechanisms of action of valproate in neuropsychiatric disorders: can we see the forest for the trees? Cell Mol Life Sci, 64, 20902103.Google Scholar
Sahraian, A, Bigdeli, M, Ghanizadeh, A, Akhondzadeh, S (2014). Topiramate as an adjuvant treatment for obsessive compulsive symptoms in patients with bipolar disorder: a randomized double blind placebo controlled clinical trial. J Affect Disord, 166, 201205.Google Scholar
Sandson, NB, Marcucci, C, Bourke, DL, Smith-Lamacchia, R (2006). An interaction between aspirin and valproate: the relevance of plasma protein displacement drug-drug interactions. Am J Psychiatry, 163, 18911896.Google Scholar
Schule, C, Baghai, TC, Eser, D, Nothdurfter, C, Rupprecht, R (2009). Lithium but not carbamazepine augments antidepressant efficacy of mirtazapine in unipolar depression: an open-label study. World J Biol Psychiatry, 10, 390399.Google Scholar
Seo, HJ, Chiesa, A, Lee, SJ, et al. (2011). Safety and tolerability of lamotrigine: results from 12 placebo-controlled clinical trials and clinical implications. Clin Neuropharmacol, 34, 3947.Google Scholar
Serrani Azcurra, DJ (2013). Lamotrigine rechallenge after a skin rash. A combined study of open cases and a meta-analysis. Rev Psiquiatr Salud Ment, 6, 144149.Google Scholar
Sie, M (2014). Mood stabilisers in the management of bipolar affective disorder. Prog Neurol Psychiatry, 18, 2232.Google Scholar
Smith, LA, Cornelius, VR, Azorin, JM, et al. (2010). Valproate for the treatment of acute bipolar depression: systematic review and meta-analysis. J Affect Disord, 122, 19.Google Scholar
Solmi, M, Veronese, N, Zaninotto, L, et al. (2016). Lamotrigine compared to placebo and other agents with antidepressant activity in patients with unipolar and bipolar depression: a comprehensive meta-analysis of efficacy and safety outcomes in short-term trials. CNS Spectr, 21, 403418.Google Scholar
Stoffers, J, Vollm, BA, Rucker, G, et al. (2010). Pharmacological interventions for borderline personality disorder. Cochrane Database Syst Rev, (6), CD005653.Google Scholar
Takezaki, H, Hanaoka, M (1971). The use of carbamazepine (Tegretol) in the control of manic depressive psychosis and other manic depressive states. Saishin Igaku, 13, 13101318 (in Japanese).Google Scholar
Tanoshima, M, Kobayashi, T, Tanoshima, R, et al. (2015). Risks of congenital malformations in offspring exposed to valproic acid in utero: a systematic review and cumulative meta-analysis. Clin Pharmacol Ther, 98, 417441.Google Scholar
Taylor, DM, Cornelius, V, Smith, L, Young, AH (2014). Comparative efficacy and acceptability of drug treatments for bipolar depression: a multiple-treatments meta-analysis. Acta Psychiatr Scand, 130, 452469.Google Scholar
Taylor, D, Paton, C, Kapur, S (2015). The Maudsley Prescribing Guidelines in Psychiatry, 12th ed. Hoboken: Wiley Blackwell.Google Scholar
Thomas, SV, Ajaykumar, B, Sindhu, K, et al. (2008). Motor and mental development of infants exposed to antiepileptic drugs in utero. Epilepsy Behav, 13, 229236.Google Scholar
Thorn, CF, Leckband, SG, Kelsoe, J, et al. (2011). PharmGKB summary: carbamazepine pathway. Pharmacogenet Genomics, 21, 906910.Google Scholar
Tomson, T, Battino, D, Perucca, E (2016). Valproic acid after five decades of use in epilepsy: time to reconsider the indications of a time-honoured drug. Lancet Neurol, 15, 210218.Google Scholar
Tseng, PT, Chen, YW, Chung, W, et al. (2016). Significant effect of valproate augmentation therapy in patients with schizophrenia: a meta-analysis study. Medicine, 95, e2475.Google Scholar
Uguz, F, Sharma, V (2016). Mood stabilizers during breastfeeding: a systematic review of the recent literature. Bipolar Disord, 18, 325333.Google Scholar
Vajda, FJ, Donnan, GA, Phillips, J, Bladin, PF (1981). Human brain, plasma, and cerebrospinal fluid concentration of sodium valproate after 72 hours of therapy. Neurology, 31, 486487.Google Scholar
Vajda, FJ, O’Brien, TJ, Graham, JE, Lander, CM, Eadie, MJ (2013). Dose dependence of fetal malformations associated with valproate. Neurology, 81, 9991003.Google Scholar
Vasudev, K, Keown, P, Gibb, I, McAllister-Williams, RH (2010). Hematological effects of valproate in psychiatric patients: what are the risk factors? J Clin Psychopharmacol, 30(3), 282285.Google Scholar
van der Loos, ML, Mulder, PG, Hartong, EG, et al.; LamLit Study Group (2009). Efficacy and safety of lamotrigine as add-on treatment to lithium in bipolar depression: a multicenter, double-blind, placebo-controlled trial. J Clin Psychiatry, 70, 223231.Google Scholar
Vella, T, Mifsud, J (2014). Interactions between valproic acid and quetiapine/olanzapine in the treatment of bipolar disorder and the role of therapeutic drug monitoring. J Pharmacy Pharmacol, 66, 747759.Google Scholar
Veroniki, AA, Cogo, E, Rios, P, et al. (2017a). Comparative safety of anti-epileptic drugs during pregnancy: a systematic review and network meta-analysis of congenital malformations and prenatal outcomes. BMC Med, 15, 95.Google Scholar
Veroniki, AA, Rios, P, Cogo, E, et al. (2017b). Comparative safety of antiepileptic drugs for neurological development in children exposed during pregnancy and breast feeding: a systematic review and network meta-analysis. BMJ Open, 7, e017248.Google Scholar
Vieta, E, Manuel Goikolea, J, Martinez-Aran, A, et al. (2006). A double-blind, randomized, placebo-controlled, prophylaxis study of adjunctive gabapentin for bipolar disorder. J Clin Psychiatry, 67, 473477.Google Scholar
Wagner, KD, Kowatch, RA, Emslie, GJ, et al. (2006). A double-blind, randomized, placebo-controlled trial of oxcarbazepine in the treatment of bipolar disorder in children and adolescents. Am J Psychiatry, 163, 11791186. Erratum in: Am J Psychiatry 2006, 163(10), 1843.Google Scholar
Wang, Y, Xia, J, Helfer, B, Li, C, Leucht, S (2008). Valproate for schizophrenia. Cochrane Database Syst Rev, (3), CD004028.Google Scholar
Wegner, C, Nau, H (1992). Alteration of embryonic folate metabolism by valproic acid during organogenesis: implications for mechanism of teratogenesis. Neurology, 42, 1724.Google Scholar
Weisler, RH, Calabrese, JR, Bowden, CL, et al. (2008). Discovery and development of lamotrigine for bipolar disorder: a story of serendipity, clinical observations, risk taking, and persistence. J Affect Disord, 108, 19.Google Scholar
Wieck, A, Jones, S (2018). Dangers of valproate in pregnancy. BMJ, 361, k1609.Google Scholar
Yasam, VR, Jakki, SL, Senthil, V, et al. (2016). A pharmacological overview of lamotrigine for the treatment of epilepsy. Expert Rev Clin Pharmacol, 9, 15331546.Google Scholar
Yatham, LN, Kennedy, SH, Parikh, SV, et al. (2018). Canadian Network for Mood and Anxiety Treatments (CANMAT) and International Society for Bipolar Disorders (ISBD) 2018 guidelines for the management of patients with bipolar disorder. Bipolar Disord, 20, 97170.Google Scholar
Yildiz, A, Nikodem, M, Vieta, E, Correll, CU, Baldessarini, RJ (2015). A network meta-analysis on comparative efficacy and all-cause discontinuation of antimanic treatments in acute bipolar mania. Psychol Med, 45, 299317.Google Scholar
Zhang, L, Li, H, Li, S, Zou, X (2016). Reproductive and metabolic abnormalities in women taking valproate for bipolar disorder: a meta-analysis. Eur J Obstet Gynecol Reprod Biol, 202, 2631.Google Scholar
Zhang, ZJ, Tan, QR, Tong, Y, et al. (2008). The effectiveness of carbamazepine in unipolar depression: a double-blind, randomized, placebo-controlled study. J Affect Disord, 109, 9197.Google Scholar
Zhou, X, Ravindran, AV, Qin, B, et al. (2015). Comparative efficacy, acceptability, and tolerability of augmentation agents in treatment-resistant depression: systematic review and network meta-analysis. J Clin Psychiatry, 76, e487e498.Google Scholar

References

Adler, L, Wilen, T, Zhang, S, et al. (2009). Retrospective safety analysis of atomoxetine in adult ADHD patients with or without comorbid alcohol abuse and dependence. Am J Addict, 18(5), 393401.Google Scholar
Advokat, C (2010). What are the cognitive effects of stimulant medications? Emphasis on adults with attention-deficit/hyperactivity disorder (ADHD). Neurosci Biobehav Rev, 34(8), 12551266.Google Scholar
Advokat, C, Lane, SM, Luo, C (2011). College students with and without ADHD: comparison of self-report of medication usage, study habits, and academic achievement. J Atten Disord, 15, 656666.Google Scholar
Ameis, SH, Szatmari, P (2012). Imaging-genetics in autism spectrum disorder: advances, translational impact and future directions. Front Psychiatry, 3(46), 113.Google Scholar
Ameis, SH, Kassee, C, Corbett-Dick, P, et al. (2018). Systematic review and guide to management of core and psychiatric symptoms in youth with autism. Acta Psychiatr Scand, 138, 379400.Google Scholar
American Psychiatric Association (APA) (1980). Diagnostic and Statistical Manual of Mental Disorders, 3rd ed. Arlington, VA: American Psychiatric Association.Google Scholar
American Psychiatric Association (APA) (2013). Diagnostic and Statistical Manual of Mental Disorders, 5th ed. Arlington, VA: American Psychiatric Association.Google Scholar
Andrew, BN, Guan, NC, Jaafar, NRN (2018). The use of methylphenidate for physical and psychological symptoms in cancer patients: a review. Curr Drug Targets, 19(8), 877887.Google Scholar
Asherson, P, Young, AH, Eich-Höchli, D, et al. (2014). Differential diagnosis, comorbidity and treatment of attention-deficit/hyperactivity disorder in relation to bipolar disorder or borderline personality disorder in adults. Curr Med Res Opin, 30(8), 16571672.Google Scholar
Biederman, J, Lindsten, A, Sluth, LB, et al. (2019). Vortioxetine for attention deficit hyperactivity disorder in adults: a randomized, double-blind, placebo-controlled, proof-of-concept study. J Psychopharmacol, 33(4), 511521.Google Scholar
Bijlenga, D, Kulcu, S, van Gellecum, T, Eryigit, Z, Kooij, JJS (2017). Persistence and adherence to psychostimulants, and psychological well-being up to 3 years after specialized treatment of adult attention-deficit/hyperactivity disorder: a naturalistic follow-up study. J Clin Psychopharmacol, 37(6), 689696.Google Scholar
Boilson, M, Shah, P and Royal College of Psychiatrists in Scotland special interest group in ADHD (2017). ADHD in Adults: Good Practice Guidelines, London: The Royal College of Psychiatrists.Google Scholar
Bolea-Alamanac, BM, Green, A, Verma, G, Maxwell, P, Davies, SJC (2013). Methylphenidate use in pregnancy and lactation: a systematic review of evidence. Br J Clin Pharmacol, 77(1), 96101.Google Scholar
Bolea-Alamañac, B, Nutt, DJ, Adamou, M, et al. (2014). Evidence-based guidelines for the pharmacological management of attention deficit hyperactivity disorder: update on recommendations from the British Association for Psychopharmacology. J Psychopharmacol, 28(3), 179203.Google Scholar
Bonell, CP, Hickson, FC, Weatherburn, P, Reid, DS (2010). Methamphetamine use among gay men across the UK. Int J Drug Policy, 21(3), 244246.Google Scholar
Bourne, A, Reid, D, Hickson, F, Torres-Rueda, S, Weatherburn, P (2015). Illicit drug use in sexual settings (‘chemsex’) and HIV/STI transmission risk behaviour among gay men in south London: findings from a qualitative study. Sex Transm Infect, 91(8), 564568.Google Scholar
Brook, DW, Brook, JS, Zhang, C, Koppel, J (2010). Association between attention-deficit/hyperactivity disorder in adolescence and substance use disorders in adulthood. Arch Pediatr Adolesc Med, 164(10), 930934.Google Scholar
Butterfield, ME, Saal, J, Young, B, Young, JL (2016). Supplementary guanfacine hydrochloride as a treatment of attention deficit hyperactivity disorder in adults: a double blind, placebo-controlled study. Psychiatry Res, 236, 136141.Google Scholar
Caisley, H, Müller, U (2012). Adherence to medication in adults with attention deficit hyperactivity disorder and pro re nata dosing of psychostimulants: a systematic review. Eur Psychiatry, 27, 343349.Google Scholar
Canadian ADHD Resource Alliance (CADDRA) (2018). Canadian ADHD Practice Guidelines, 4th ed. Toronto: CADDRA. Available at: www.caddra.ca/canadian-adhd-practice-guidelines (last accessed 15.8.18).Google Scholar
Castaldi, S, Gelatti, U, Orizio, G, et al. (2012). Use of cognitive enhancement medication among Northern Italian university students. J Addict Med, 6, 112117.Google Scholar
Caye, A, Swanson, JM, Coghill, D, Rohde, LA (2019). Treatment strategies for ADHD: an evidence-based guide to select optimal treatment. Mol Psychiatry, 24, 390408.Google Scholar
Cederström, C (2016). Like it or not ‘smart drugs’ are coming to the office. Harvard Business Review. Available at: https://hbr.org/2016/05/like-it-or-not-smart-drugs-are-coming-to-the-office (last accessed 1.11.18).Google Scholar
Centers for Disease Control and Prevention (CDC) (2007). Methamphetamine Use and Risk for HIV/AIDS. Available at: file:///C:/Users/K1517101/Downloads/cdc_11778_DS1.pdf (last accessed 1.8.18).Google Scholar
Chan, E, Fogler, JM, Hammerness, PG (2016). Treatment of attention-deficit/hyperactivity disorder in adolescents: a systematic review. JAMA, 315, 19972008.Google Scholar
Chang, Z, Lichtenstein, P, D’Onofrio, BM, Sjölander, A, Larsson, H (2014). Serious transport accidents in adults with attention-deficit/hyperactivity disorder and the effect of medication: a population-based study. JAMA Psychiatry, 71, 319325.Google Scholar
Chang, Z, D’Onofrio, BM, Quinn, PD, Lichtenstein, P, Larsson, H (2016). Medication for attention-deficit/hyperactivity disorder and risk for depression: a nationwide longitudinal cohort study. Biol Psychiatry, 80, 916922.Google Scholar
Chang, Z, Ghirardi, L, Quinn, PD, et al. (2019). Risks and benefits of ADHD medication on behavioral and neuropsychiatric outcomes: a qualitative review of pharmacoepidemiology studies using linked prescription databases. Biol Psychiatry [Epub ahead of print] doi:doi.org/10.1016/j.biopsych.2019.04.009.Google Scholar
Chavez, C, Hollaus, M, Scarr, E, et al. (2010). The effect of estrogen on dopamine and serotonin receptor and transporter levels in the brain: an autoradiography study. Brain Res, 1321, 5159.Google Scholar
Chen, Q, Hartman, CA, Haavik, J, et al. (2018). Common psychiatric and metabolic comorbidity of adult attention-deficit/hyperactivity disorder: a population-based cross-sectional study PLoS One, 13(9), e0204516.Google Scholar
Coghill, D, Chen, W, Silva, D (2019). Organizing and delivering treatment for ADHD. In Rohde, LA, Buitelaar, JK, Gerlach, M, Faraone, SV, eds., The World Federation of ADHD Guide, pp. 93–109. [open access pdf and e-book: http://cpo-media.net/ADHD/2019/ebook/HTML/93/].Google Scholar
Cooper, RE, Williams, E, Seegobin, S, et al. (2017). Cannabinoids in attention-deficit/hyperactivity disorder: a randomised-controlled trial. Eur Neuropsychopharmacol, 27, 795808.Google Scholar
Cope, LC, Abuzour, AS, Tully, MP (2016). Nonmedical prescribing: where are we now? Ther Adv Drug Saf, 7(4), 165172.Google Scholar
Cortese, S (2018). Are the effects of methylphenidate uncertain? Ir J Psychol Med, 35, 163167.Google Scholar
Cortese, S, Tessari, L (2017). Attention-deficit/hyperactivity disorder (ADHD) and obesity: update 2016. Curr Psychiatry Rep, 19(1), 4.Google Scholar
Cortese, S, Brown, TE, Corkum, P, et al. (2013). Assessment and management of sleep problems in youths with attention-deficit/hyperactivity disorder. J Am Acad Child Adolesc Psychiatry, 52, 784796.Google Scholar
Cortese, S, D’Acunto, G, Konofal, E, Masi, G, Vitiello, B (2017). New formulations of methylphenidate for the treatment of attention-deficit/hyperactivity disorder: pharmacokinetics, efficacy, and tolerability. CNS Drugs, 31, 149160.Google Scholar
Cortese, S, Adamo, N, Del Giovane, C, et al. (2018). Comparative efficacy and tolerability of medications for attention-deficit hyperactivity disorder in children, adolescents, and adults: a systematic review and network meta-analysis. Lancet Psychiatry, 5, 727738.Google Scholar
Coulter, A, Collins, A (2011). Making shared decision-making a reality: no decision about me, without me. London: The King’s Fund.Google Scholar
Crunelle, CL, van den Brink, W, Moggi, F, et al.; ICASA consensus group, Matthys, F (2018). International consensus statement on screening, diagnosis and treatment of substance use disorder patients with comorbid attention deficit/hyperactivity disorder. Eur Addict Res, 24(1), 4351.Google Scholar
Cunill, R, Castells, X, Tobias, A, Capellà, D (2015). Pharmacological treatment of attention deficit hyperactivity disorder with co-morbid drug dependence. J Psychopharmacol, 29, 1523.Google Scholar
Dalley, JW, Roiser, JP (2012). Dopamine, serotonin and impulsivity. Neuroscience, 215, 4258.Google Scholar
De Crescenzo, F, Cortese, S, Adamo, N, Janiri, L (2017). Pharmacological and non-pharmacological treatment of adults with ADHD: a meta-review. Evid Based Ment Health, 20, 411.Google Scholar
DeSaints, AD, Webb, EM, Noar, SM (2008). Illicit use of prescription ADHD medications on a college campus: a multimethodological approach. J Am Coll Health, 75, 315324.Google Scholar
Diav-Citrin, O, Shechtman, S, Arnon, J, et al. (2016). Methylphenidate in pregnancy: a multicentre, prospective, comparative, observational study. J Clin Psychiatry, 77(9), 11761181.Google Scholar
Díaz-Román, A, Mitchell, R, Cortese, S (2018). Sleep in adults with ADHD: systematic review and meta-analysis of subjective and objective studies. Neurosci Biobehav Rev, 89, 6171.Google Scholar
Dietz, P, Soyka, M, Franke, AG (2016). Pharmacological neuroenhancement in the field of economics: poll results from an online survey. Front Psychol, 7(520), 18.Google Scholar
Driver & Vehicle Licensing Agency (DVLA) (2019). Assessing fitness to drive: a guide for medical professionals. Available at: www.gov.uk/government/publications/ assessing-fitness-to-drive-a-guide-for-medical-professionals (last accessed 1.04.19).Google Scholar
DuPaul, GJ, Weyandt, LL, Rossi, JS, et al. (2012). Double-bind, placebo-controlled, crossover study of the efficacy and safety of lisdexamfetamine dimesylate in college students with ADHD. J Atten Disord, 16, 202220.Google Scholar
Eakle, R, Mbogua, J, Mutanha, N, Ress, H (2018). Exploring acceptability of oral PrEP prior to implementation among female sex workers in South Africa. J Int AIDS Soc, 21, e25081.Google Scholar
Edvinsson, D, Ekselius, L (2018a). Long-term tolerability and safety of pharmacological treatment of adult attention-deficit/hyperactivity disorder: a 6-year prospective naturalistic study. J Clin Psychopharmacol, 38(4), 370375.Google Scholar
Edvinsson, D, Ekselius, L (2018b). Six-year outcome in subjects diagnosed with attention-deficit/hyperactivity disorder as adults. Eur Arch Psychiatry Clin Neurosci, 268(4), 337347.Google Scholar
Faraone, SV (2018). The pharmacology of amphetamine and methylphenidate: relevance to the neurobiology of attention-deficit/hyperactivity disorder and other psychiatric comorbidities. Neurosci Biobehav Rev, 87, 255270.Google Scholar
Findling, RL, Adler, LA, Spencer, TJ, et al. (2019). Dasotraline in children with attention-deficit/hyperactivity disorder: a six-week, placebo-controlled, fixed-dose trial. J Child Adolesc Psychopharmacol, 29(2), 8089.Google Scholar
Fredriksen, M, Dahl, AA, Martinsen, EW, et al. (2014). Effectiveness of one-year pharmacological treatment of adult attention-deficit/hyperactivity disorder (ADHD): an open-label prospective study of time in treatment, dose, side-effects and comorbidity. Eur Neuropsychopharmacol, 24, 18731884.Google Scholar
Galbraith, N (2015). The methamphetamine problem. BJPsych Bull, 39, 218220.Google Scholar
Gillberg, C, Gillberg, IC, Rasmussen, P, et al. (2004). Co-existing disorders in ADHD: implications for diagnosis and intervention. Eur Child Adolesc Psychiatry, 13(Suppl 1), 8092.Google Scholar
Gillies, GE, McArthur, S (2010). Estrogen actions in the brain and the basis for differential action in men and women: a case for sex-specific medicines. Pharmacol Rev, 62(2), 155198.Google Scholar
Ginsberg, Y, Lindefors, N (2012). Methylphenidate treatment of adult male prison inmates with attention-deficit hyperactivity disorder: randomised double-blind placebo-controlled trial with open-label extension. Br J Psychiatry, 200(1), 6873.Google Scholar
Golubchik, P, Sever, J, Zalsman, G, Weizman, A (2008). Methylphenidate in the treatment of female adolescents with cooccurrence of attention deficit/hyperactivity disorder and borderline personality disorder: a preliminary open-label trial. Int Clin Psychopharmacol, 23(4), 228231.Google Scholar
Gonzales, R, Mooney, L, Rawson, RA (2010). The methamphetamine problem in the United States. Annu Rev Public Health, 31, 385398.Google Scholar
Gould, ON, Doucette, C (2018). Self-management of adherence to prescribed stimulants in college students with ADD/ADHD. J Atten Disord, 22, 349355.Google Scholar
Graham, J, Coghill, D (2008). Adverse effects of pharmacotherapies for attention-deficit hyperactivity disorder: epidemiology, prevention and management. CNS Drugs, 22(3), 213237.Google Scholar
Graham, J, Banaschewski, T, Buitelaar, J, et al. (2011). European guidelines on managing adverse effects of medication for ADHD. Eur Child Adolesc Psychiatry, 20(1), 1737.Google Scholar
Greely, H, Sanakian, B, Harris, J, et al. (2008). Towards responsible use of cognitive-enhancing drugs by the healthy. Nature, 456, 702705.Google Scholar
Halkitis, PN (2010). Reframing HIV prevention for gay men in the United States. Am Psychol, 65(8), 752763.Google Scholar
Halkitis, PN, Levy, MD, Solomon, TM (2016). Temporal relations between methamphetamine use and HIV seroconversion in gay, bisexual, and other men who have sex with men. J Health Psychol, 21(1), 9399.Google Scholar
Halperin, JM, Marks, DJ (2019). Practitioner review: assessment and treatment of preschool children with attention-deficit/hyperactivity disorder. J Child Psychol Psychiatry [Epub ahead of print] doi:10.1111/jcpp.13014.Google Scholar
Harrison, AG, Edwards, MJ, Parker, KCH (2007). Identifying students faking ADHD: preliminary findings and strategies for detection. Arch Clin Neuropsychol, 22, 577588.Google Scholar
Heal, DJ, Smith, S, Gosden, J, Nutt, DJ (2013). Amfetamine, past and present: a pharmacological and clinical perspective. J Psychopharmacol, 27, 118.Google Scholar
Holloway, K, Bennett, T (2012). Prescription drug misuse among university staff and students: a survey of motives, nature and extent. Drug Educ Prev Polic, 19, 137144.Google Scholar
Hooberman, D, Stein, TA (1984). Treatment of attention deficit and borderline personality disorders with psychostimulants: case report. J Clin Psychiatry, 45, 441442.Google Scholar
Hornig-Rohan, M, Amsterdam, JD (2002). Venlafaxine versus stimulant therapy in patients with dual diagnosis ADD and depression. Prog Neuropsychopharmacol Biol Psychiatry, 26(3), 585589.Google Scholar
Humphreys, KL, Eng, T, Lee, SS (2013). Stimulant medication and substance use outcomes: a meta-analysis. JAMA Psychiatry, 70, 740749.Google Scholar
Husain, M, Mehta, MA (2011). Cognitive enhancement by drugs in health and disease. Trends Cogn Sci, 15(1), 2836.Google Scholar
Huss, M, McBurnett, K, Cutler, AJ, et al. (2019). Distinguishing the efficacy and sedative effects of guanfacine extended release in children and adolescents with attention-deficit/hyperactivity disorder. Eur Neuropsychopharmacol, 29, 432443.Google Scholar
Iaccarino, MA, Philpotts, LL, Zafonte, R, Biederman, J (2018). Stimulant use in the management of mild traumatic brain injury: a qualitative literature review. J Atten Disord [Epub ahead of print] doi:10.1177/1087054718759752.Google Scholar
Ioannidis, K, Chamberlain, SR, Müller, U (2014a). Ostracizing caffeine from the pharmacological arsenal for ADHD: was this a correct decision? A literature review. J Psychopharmacol, 28, 830836.Google Scholar
Ioannidis, K, Serfontein, J, Müller, U (2014b). Bulimia nervosa patient diagnosed with previously unsuspected adult ADHD: clinical case report, literature review, and diagnostic challenges. Int J Eat Disord, 47(4), 431436.Google Scholar
Iwanami, A, Saito, K, Fujiwara, M, Okutsu, D, Ichikawa, H (2019). Randomized, double-blind, placebo-controlled, phase 3 study of guanfacine extended release in adults with attention-deficit/hyperactivity disorder. Presented at 7th World Congress on ADHD, Lisbon, Portugal.Google Scholar
Jackson, CO (1971). The amphetamine inhaler: a case study of medical abuse. J Hist Med, 26, 187196.Google Scholar
Kessler, RC, Adler, L, Barkley, R, et al. (2006). The prevalence and correlates of adult ADHD in the United States: results from the National Comorbidity Survey Replication. Am J Psychiatry, 163(4), 716723.Google Scholar
Khan, A, Fahl Mar, K, Brown, WA (2017). Does the increasing placebo response impact outcomes of adult and pediatric ADHD clinical trials? Data from the US Food and Drug Administration 2000–2009. J Psychiatr Res, 94, 202207.Google Scholar
Koblan, KS, Hopkins, SC, Sarma, K, et al. (2016). Assessment of human abuse potential of dasotraline compared to methylphenidate and placebo in recreational stimulant users. Drug Alcohol Depend, 159, 2634.Google Scholar
Konstenius, M, Jayaram-Lindström, N, Guterstam, J, et al. (2014). Methylphenidate for attention deficit hyperactivity disorder and drug relapse in criminal offenders with substance dependence: a 24-week randomized placebo-controlled trial. Addiction, 109(3), 440449.Google Scholar
Kooij, JJS (2013). Adult ADHD: Diagnostic Assessment and Treatment, 3rd ed. London: Springer.Google Scholar
Kooij, JJ, Huss, M, Asherson, P, et al. (2012). Distinguishing comorbidity and successful management of adult ADHD. J Atten Disord, 16(5 Suppl), 319.Google Scholar
Kooij, JJ, Michielsen, M, Kruithof, H, Bijlenga, D (2016). ADHD in old age: a review of the literature and proposal for assessment and treatment. Expert Rev Neurother, 16(12), 13711381.Google Scholar
Kooij, JJS, Bijlenga, D, Salerno, L, et al. (2019). Updated European Consensus Statement on diagnosis and treatment of adult ADHD. Eur Psychiatry, 56, 1434.Google Scholar
Kooij, SJ, Bejerot, S, Blackwell, A, et al. (2010). European consensus statement on diagnosis and treatment of adult ADHD: the European Network Adult ADHD. BMC Psychiatry, 10, 67.Google Scholar
Kratochvil, CJ, Newcorm, JH, Arnold, LE, et al. (2005). Atomoxetine alone or combined with fluoxetine for treating ADHD with comorbid depressive or anxiety symptoms. J Am Acad Child Adolesc Psychiatry, 44(9), 915924.Google Scholar
Levin, FR, Mariani, JJ, Specker, S, et al. (2015). Extended-release mixed amphetamine salts vs placebo for comorbid adult attention-deficit/hyperactivity disorder and cocaine use disorder. JAMA Psychiatry, 72(6), 593602.Google Scholar
Levin, FR, Choi, CJ, Pavlicova, M, et al. (2018). How treatment improvement in ADHD and cocaine dependence are related to one another: a secondary analysis. Drug Alcohol Depend, 188, 135140.Google Scholar
Lichtenstein, P, Halldner, L, Zetterqvist, J, et al. (2012). Medication for attention deficit-hyperactivity disorder and criminality. N Engl J Med, 367, 20062014.Google Scholar
Lorvic, J, Bourgois, P, Wenger, LD, et al. (2012). Sexual pleasure and sexual risk among women who use methamphetamine: a mixed methods study. Int J Drug Policy, 23(5), 385392.Google Scholar
Lu, Y, Sjölander, A, Cederlöf, M, et al. (2017). Association between medication use and performance on higher education entrance tests in individuals with attention-deficit/hyperactivity disorder. JAMA Psychiatry, 74(8), 815822.Google Scholar
Magon, RK, Latheesh, B, Müller, U (2015). Specialist community adult ADHD clinics in East Anglia: service evaluation and audit of NICE guideline recommendations. BJPsych Bull, 39, 36140.Google Scholar
Maier, LJ, Liechti, ME, Herzig, F, Schaub, MP (2013). To dope or not to dope: neuroenhancement with prescription drugs and drugs of abuse among Swiss university students. PLoS One, 8(11), e77967.Google Scholar
Man, KKC, Ip, P, Chan, EW, et al. (2017). Effectiveness of pharmacological treatment for attention-deficit/hyperactivity disorder on physical injuries: a systematic review and meta-analysis of observational studies. CNS Drugs, 31, 10431055.Google Scholar
Martins, S, Tramontina, S, Polanczyk, G, et al. (2004). Weekend holidays during methylphenidate use in ADHD children: a randomized clinical trial. J Child Adolesc Psychopharmacol, 14(2), 195206.Google Scholar
Matthies, S, Philipsen, A (2016). Comorbidity of personality disorders and adult attention deficit hyperactivity disorder (ADHD): review of recent findings. Curr Psychiatry Rep, 18(4), 33.Google Scholar
McArdle, A (2016). Attention deficit hyperactivity disorder. In Currie, A, Owen, B, eds., Sports Psychiatry. Oxford: Oxford University Press, pp. 8795.Google Scholar
McIntyre, RS, Lee, Y, Zhou, AJ, et al. (2017). The efficacy of psychostimulants in major depressive episodes: a systematic review and meta-analysis. J Clin Psychopharmacol, 37, 412418.Google Scholar
Melendez-Torres, GJ, Bonell, C, Hickson, F, et al. (2016). Predictors of crystal methamphetamine use in a community-based sample of UK men who have sex with men. Int J Drug Policy, 36, 4346.Google Scholar
Moran, LV, Ongur, D, Hsu, J, et al. (2019). Psychosis with methylphenidate or amphetamine in patients with ADHD. N Engl J Med, 380, 11281138.Google Scholar
Müller, U (2008). Pharmacological treatment. In Cappa, SF, Abutalebi, J, Démonet, J-F, Fletcher, P, Garrard, P, eds., Cognitive Neurology: A Clinical Textbook. Oxford: Oxford University Press, pp. 475498.Google Scholar
Müller-Sedgwick, U, Meisel, G, Nasri, M, Sedgwick-Müller, JA (2018). How fast should we titrate methylphenidate in adults with ADHD? A meta-analysis of dose titration regimes in RCTs. presented at 5th Eunethydis International Conference on ADHD, Edinburgh, Scotland.Google Scholar
National Institute for Health and Care Excellence (NICE) (2008). Attention deficit hyperactivity disorder: diagnosis and management. Clinical guideline [CG72]. London: National Institute for Health and Care Excellence.Google Scholar
National Institute for Health and Care Excellence (NICE) (2018). Attention deficit hyperactivity disorder: diagnosis and management. NICE guideline [NG87]. London: National Institute for Health and Care Excellence.Google Scholar
Newcorn, JH, Stein, MA, Childress, AC, et al. (2013). Randomized, double-blind trial of guanfacine extended release in children with attention-deficit/hyperactivity disorder: morning or evening administration. J Am Acad Child Adolesc Psychiatry, 52, 921930.Google Scholar
NHS England (2016). The Five Year Forward View for Mental Health, Mental Health Taskforce. Available at: www.england.nhs.uk/mentalhealth/taskforce (last accessed 24.10.18).Google Scholar
Nigg, JT, Johnstone, JM, Musser, ED, et al. (2016). Attention-deficit/hyperactivity disorder (ADHD) and being overweight/obesity: new data and meta-analysis. Clin Psychol Rev, 43, 6779.Google Scholar
Nussbaum, NL (2012). ADHD and female specific concerns: a review of the literature and clinical implications, J Atten Disord, 16, 87100.Google Scholar
Nutt, DJ, Fone, K, Asherson, P, et al.; British Association for Psychopharmacology (2007). Evidence-based guidelines for management of attention-deficit/hyperactivity disorder in adolescents in transition to adult services and in adults: recommendations from the British Association for Psychopharmacology. J Psychopharmacol, 21(1), 1041.Google Scholar
Ogundele, MO, Ayyash, HF (2018). Review of the evidence for the management of co-morbid tic disorders in children and adolescents with attention deficit hyperactivity disorder. World J Clin Pediatr, 7, 3642.Google Scholar
Ott, R, Biller-Andorno, N (2014). Neuroenhancement among Swiss students: a comparison of users and non-users. Pharmacopsychiatry, 47, 2228.Google Scholar
Padala, PR, Padala, KP, Lensing, SY, et al. (2018). Methylphenidate for apathy in community-dwelling older veterans with mild Alzheimer’s disease: a double-blind, randomized, placebo-controlled trial. Am J Psychiatry, 175, 159168.Google Scholar
Park, J, Choi, HW, Yum, MS, et al. (2018). Relationship between aggravation of seizures and methylphenidate treatment in subjects with attention-deficit/hyperactivity disorder and epilepsy. J Child Adolesc Psychopharmacol, 28, 537546.Google Scholar
Perera, B (2018). Attention deficit hyperactivity disorder in people with intellectual disability. Ir J Psychol Med, 35, 213219.Google Scholar
Philipsen, A, Richter, H, Peters, J, et al. (2007). Structured group psychotherapy in adults with attention deficit hyperactivity disorder: results of an open multicentre study. J Nerv Ment Disord, 195(12), 10131019.Google Scholar
Philipsen, A, Limberger, MF, Lieb, K, et al. (2008). Attention-deficit hyperactivity disorder as a potentially aggravating factor in borderline personality disorder. Br J Psychiatry, 192(2), 118123.Google Scholar
Philipsen, A, Jans, T, Graf, E, et al.; Comparison of Methylphenidate and Psychotherapy in Adult ADHD Study (COMPAS) Consortium (2015). Effects of group psychotherapy, individual counseling, methylphenidate, and placebo in the treatment of adult attention-deficit/hyperactivity disorder: a randomized clinical trial. JAMA Psychiatry, 72, 11991210.Google Scholar
POMH-UK (2015). Topic 13b re-audit report. Prescribing for ADHD in children, adolescents and adults. Prescribing Observatory for Mental Health (POMH), CCQI218 (data on file).Google Scholar
Quinn, PO (2005). Treating adolescent girls and women with ADHD: gender-specific issue. J Clin Psychol, 61(5), 579587.Google Scholar
Rabiner, DL, Anastopoulos, AD, Costello, EJ, et al. (2009). The misuse and diversion of prescribed ADHD medications by college students. J Atten Disord, 13, 144153.Google Scholar
Renoux, C, Shin, JY, Dell’Aniello, S, Fergusson, E, Suissa, S (2016). Prescribing trends of attention-deficit hyperactivity disorder (ADHD) medications in UK primary care, 1995–2015. Br J Clin Pharmacol, 82, 858868.Google Scholar
Reyniers, T, Hoornenborg, E, Vuylsteke, B, Wouters, K, Laga, M (2017). Pre-exposure prophylaxis (PrEP) for men who have sex with men in Europe: review of evidence for a much needed prevention tool. Sex Transm Infect, 93, 363367.Google Scholar
Rösler, M, Fischer, R, Ammer, R, Ose, C, Retz, W (2009). A randomised, placebo-controlled, 24-week, study of low-dose extended-release methylphenidate in adults with attention-deficit/hyperactivity disorder. Eur Arch Psychiatry Clin Neurosci, 259(2), 120129.Google Scholar
Roy, A, Hechtman, L, Arnold, LE, et al.; MTA Cooperative Group (2017). Childhood predictors of adult functional outcomes in the multimodal treatment study of attention-deficit/hyperactivity disorder (MTA). J Am Acad Child Adolesc Psychiatry, 56(8), 687695.Google Scholar
Royal College of Psychiatrists (RCPsych) Psychopharmacology Committee (2017). Use of Licensed Medicines for Unlicensed Applications in Psychiatric Practice, 2nd ed. College Report CR210. London: Royal College of Psychiatrists. Available at: www.bap.org.uk/pdfs/CR210-December2017.pdf (last accessed 8.8.19).Google Scholar
Ruggiero, S, Clavenna, A, Reale, L, et al. (2014). Guanfacine for attention deficit and hyperactivity disorder in pediatrics: a systematic review and meta-analysis. Eur Neuropsychopharmacol, 24(10), 15781590.Google Scholar
Ruthirakuhan, MT, Herrmann, N, Abraham, EH, Chan, S, Lanctôt, KL (2018). Pharmacological interventions for apathy in Alzheimer’s disease. Cochrane Database Syst Rev, (5), CD012197.Google Scholar
Sahakian, BJ, Morein-Zamir, S (2015). Pharmacological cognitive enhancement: treatment of neuropsychiatric disorders and lifestyle use by healthy people. Lancet Psychiatry, 2, 357362.Google Scholar
Scheffer, RE (2007). Concurrent ADHD and bipolar disorder. Curr Psychiatry Rep, 9, 415419.Google Scholar
Scheffer, RE, Kowatch, RA, Carmody, T, Rush, AJ (2005). Randomized, placebo-controlled trial of mixed amphetamine salts for symptoms of comorbid ADHD in pediatric bipolar disorder after mood stabilization with divalproex sodium. Am J Psychiatry, 162(1), 5864.Google Scholar
Sedgwick, JA (2018). University students with attention deficit hyperactivity disorder (ADHD): a literature review. Ir J Psychol Med, 35(3), 221235.Google Scholar
Seixas, M, Weiss, M, Müller, U (2012). Systematic review of national and international guidelines on attention deficit hyperactivity disorder (ADHD). J Psychopharmacology, 26(6), 753765.Google Scholar
Shanmugan, S, Epperson, CN (2014). Estrogen and the prefrontal cortex: towards a new understanding of estrogen’s effects on executive functions in the menopause transition. Hum Brain Mapp, 35(3), 847865.Google Scholar
Singh, I, Bard, I, Jackson, J (2014). Robust resilience and substantial interest: a survey of pharmacological cognitive enhancement among university students in the UK and Ireland. PLoS One, 9(20), e105969.Google Scholar
Slade, M (2017). Implementing shared decision making in routine mental health care. World Psychiatry, 16(2), 146153.Google Scholar
Solberg, BS, Haavik, J, Halmøy, A (2019). Health care services for adults with ADHD: patient satisfaction and the role of psycho-education. J Atten Disord, 23, 99108.Google Scholar
Solmi, M, Fornaro, M, Toyoshima, K, et al. (2018). Systematic review and exploratory meta-analysis of the efficacy, safety, and biological effects of psychostimulants and atomoxetine in patients with schizophrenia or schizoaffective disorder. CNS Spectr [Epub ahead of print] doi:10.1017/S1092852918001050.Google Scholar
Spruyt, K, Gozal, D (2011). Sleep disturbances in children with attention-deficit/hyperactivity disorder. Expert Rev Neurother, 11, 565577.Google Scholar
Stein, M, Thurmond, P, Bailey, G (2014). Willingness to use HIV pre-exposure prophylaxis among opiate users. AIDS Behav, 18(9), 16941700.Google Scholar
Stoffers, JM, Völlm, BA, Rücker, G, et al. (2012). Psychological therapies for people with borderline personality disorder. Cochrane Database Syst Rev, (8), CD005652.Google Scholar
Sugden, C, Housden, CR, Aggarwal, R, Sahakian, BJ, Darzi, A (2012). Effect of pharmacological enhancement on the cognitive and clinical psychomotor performance of sleep-deprived doctors: a randomized controlled trial. Ann Surg, 255(2), 222227.Google Scholar
Surman, CBH, Fried, R, Rhodewalt, L, Boland, H (2017). Do pharmaceuticals improve driving in individuals with ADHD? A review of the literature and evidence for clinical practice. CNS Drugs, 31, 857866.Google Scholar
Svedlund, NE, Norring, C, Ginsberg, Y, von Hausswolff-Juhlin, Y (2018). Are treatment results for eating disorders affected by ADHD symptoms? A one-year follow-up of adult females. Eur Eat Disord Rev, 26, 337345.Google Scholar
Swanson, JM, Arnold, LE, Molina, BSG, et al.; MTA Cooperative Group (2017). Young adult outcomes in the follow-up of the multimodal treatment study of attention-deficit/hyperactivity disorder: symptom persistence, source discrepancy, and height suppression. J Child Psychol Psychiatry, 58(6), 663678.Google Scholar
Tarrant, N, Roy, M, Deb, S, et al. (2018). The effectiveness of methylphenidate in the management of attention deficit hyperactivity disorder (ADHD) in people with intellectual disabilities: a systematic review. Res Dev Disabil, 83, 217232.Google Scholar
Taylor, E (2017). Attention deficit hyperactivity disorder: overdiagnosed or diagnoses missed? Arch Dis Child, 102, 376379.Google Scholar
Taylor, E (2019). ADHD medication in the longer term. Z Kinder Jugendpsychiatr Psychother [Epub ahead of print] doi:10.1024/1422-4917/a000664.Google Scholar
Thorpy, MJ (2015). Update on therapy for narcolepsy. Curr Treat Options Neurol, 17(5), 347.Google Scholar
Tomlinson, D, Robinson, PD, Oberoi, S, et al. (2018). Pharmacologic interventions for fatigue in cancer and transplantation: a meta-analysis. Curr Oncol, 25, e152e167.Google Scholar
Torgersen, T, Gjervan, B, Lensing, MB, Rasmussen, K (2016). Optimal management of ADHD in older adults. Neuropsychiatr DisTreat, 12, 7987.Google Scholar
Tritsch, NX, Sabatini, BL (2012). Dopaminergic modulation of synaptic transmission in cortex and striatum. Neuron, 76(1), 3350.Google Scholar
UK Medicines Information (2017). Which medicines should be considered for brand-name prescribing in primary care? Available at: www.sps.nhs.uk/wp-content/uploads/2017/12/UKMi_QA_Brand-name_prescribing_Update_Nov2017.pdf (last accessed 9.8.19).Google Scholar
van Reekum, R, Links, PS (1994). N of 1 study: methylphenidate in a patient with borderline personality disorder and attention deficit hyperactivity disorder. Can J Psychiatry, 39(3), 186187.Google Scholar
Verrotti, A, Moavero, R, Panzarino, G, et al. (2018). The challenge of pharmacotherapy in children and adolescents with epilepsy-ADHD comorbidity. Clin Drug Investig, 38(1), 18.Google Scholar
Viktorin, A, Rydén, E, Thase, ME, et al. (2017). The risk of treatment-emergent mania with methylphenidate in bipolar disorder. Am J Psychiatry, 174(4), 341348.Google Scholar
Wilens, TE, Morrison, NR (2011). The intersection of attention-deficit/hyperactivity disorder and substance abuse. Curr Opin Psychiatry, 24(4), 280285.Google Scholar
Wilens, TE, Adler, LA, Adams, J, et al. (2008). Misuse and diversion of stimulants prescribed for ADHD: a systematic review of the literature. J Am Acad Child Adolesc Psychiatry, 47, 2131.Google Scholar
Wilens, TE, Adler, LA, Weiss, MD, et al.; Atomoxetine ADHD/SUD Study Group (2008). Atomoxetine treatment of adults with ADHD and comorbid alcohol use disorders. Drug Alcohol Depend, 96(12), 145154.Google Scholar
Williamson, D, Johnston, C (2015). Gender differences in adults with attention-deficit/hyperactivity disorder: a narrative review. Clin Psychol Rev, 40, 1527.Google Scholar
Wilson, SJ, Nutt, DJ, Alford, C, et al. (2010). British Association for Psychopharmacology consensus statement on evidence-based treatment of insomnia, parasomnias and circadian rhythm disorders. J Psychopharmacol, 24, 15771601.Google Scholar
Wolraich, M, Brown, L, Brown, RT, et al. (2011). ADHD: clinical practice guideline for the diagnosis, evaluation and treatment of attention-deficit/hyperactivity disorder in children and adolescents. Pediatrics, 128(5), 10071022.Google Scholar
Wood, DR, Reimherr, FW, Wender, PH, Johnson, GE (1976). Diagnosis and treatment of minimal brain dysfunction in adults: a preliminary report. Arch Gen Psychiatry, 33(12), 14531460.Google Scholar
World Health Organization (2011). Technical Brief 1: Patterns and consequences of the use of amphetamine-type stimulants (ATS). Available at: www.wpro.who.int/hiv/documents/docs/Brief1forweb_850A.pdf (last accessed 1.8.18).Google Scholar

References

Aisen, P (2017). Continuing progress in Alzheimer’s disease trials. J Prev Alz Dis, 4(4), 211212.Google Scholar
Auriacombe, S, Pere, J-J, Loria-Kanza, Y, Vellas, B (2002). Efficacy and safety of rivastigmine in patients with Alzheimer’s disease who failed to benefit from treatment with donepezil. Curr Med Res Opin, 18, 129138. doi:10.1185/030079902125000471.Google Scholar
Banerjee, S, Hellier, J, Romeo, R, et al. (2013). Study of the use of antidepressants for depression in dementia: the HTA-SADD trial – a multicentre, randomised, double-blind, placebo-controlled trial of the clinical effectiveness and cost-effectiveness of sertraline and mirtazapine. Health Technol Assess, 17, 1166.Google Scholar
Beck, AT, Rush, A, Shaw, B, Emery, G (1979). Cognitive Therapy of Depression. New York: Guilford Press.Google Scholar
Bickel, U, Thomsen, T, Weber, W, et al. (1991). Pharmacokinetics of galanthamine in humans and corresponding cholinesterase inhibition. Clin Pharmacol Ther, 50, 420428.Google Scholar
Birks, J, Grimley Evans, J, Iakovidou, V, Tsolaki, M (2009). Rivastigmine for Alzheimer’s disease. Cochrane Database Syst Rev, (2), CD001191. doi:10.1002/14651858.CD001191.pub2.Google Scholar
Bolea-Alamanac, BM, Davies, SJ, Christmas, DM, et al. (2011). Cyproterone to treat aggressivity in dementia: a clinical case and systematic review. J Psychopharmacol, 25(1), 141145.Google Scholar
Braak, H, Del Tredici, K (2015). Neuroanatomy and pathology of sporadic Alzheimer’s disease. Adv Anat Embryol Cell Biol, 215, 1162.Google Scholar
Braak, H, Ghebremedhin, E, Rüb, U, Bratzke, H, Del Tredici, K (2004). Stages in the development of Parkinson’s disease-related pathology. Cell Tissue Res, 318, 121134.Google Scholar
Bullock, R, Connolly, C (2002). Switching cholinesterase inhibitor therapy in Alzheimer’s disease: donepezil to rivastigmine, is it worth it? Int J Geriatr Psychiatry, 17, 288289.Google Scholar
Campbell, NL, Perkins, AJ, Gao, S, et al. (2017). Adherence and tolerability of Alzheimer’s disease medications: a pragmatic randomized trial. J Am Geriatr Soc, 65, 14971504.Google Scholar
Chen, YD, Zhang, J, Wang, Y, Yuan, JL, Hu, WL (2016). Efficacy of cholinesterase inhibitors in vascular dementia: an updated meta-analysis. Eur Neurol, 75, 132141.Google Scholar
Chew, ML, Mulsant, BH, Pollock, BG, et al. (2008). Anticholinergic activity of 107 medications commonly used by older adults. J Am Geriatr Soc, 56, 13331341.Google Scholar
Collerton, D, Mosimann, UP, Perry, E (2015). The Neuroscience of Visual Hallucinations. Chichester: John Wiley & Sons.Google Scholar
Corbett, A, Burns, A, Ballard, C (2014). Don’t use antipsychotics routinely to treat agitation and aggression in people with dementia. BMJ, 349, g6420.Google Scholar
Cummings, JL, Mega, M, Gray, K, et al. (1994). The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementia. Neurology, 44, 23082314.Google Scholar
Cummings, JL, Geldmacher, D, Farlow, M, et al. (2013). High-dose donepezil (23 mg/day) for the treatment of moderate and severe Alzheimer’s disease: drug profile and clinical guidelines. CNS Neurosci Ther, 19, 294301.Google Scholar
Cummings, J, Ballard, C, Tariot, P, et al. (2018). Pimavanserin: potential treatment for dementia-related psychosis. J Prev Alzheimers Dis, 5(4), 253258.Google Scholar
Dawson, GR, Iversen, SD (1993). The effects of novel cholinesterase inhibitors and selective muscarinic receptor agonists in tests of reference and working memory. Behav Brain Res, 57, 143153.Google Scholar
de Vasconcelos Cunha, UG, Lopes Rocha, F, Ávila de Melo, R, et al. (2007). A placebo-controlled double-blind randomized study of venlafaxine in the treatment of depression in dementia. Dement Geriatr Cogn Disord, 24, 3641. doi:10.1159/000102570.Google Scholar
Devos, D, Moreau, C, Maltête, D, et al. (2014). Rivastigmine in apathetic but dementia and depression-free patients with Parkinson’s disease: a double-blind, placebo-controlled, randomised clinical trial. J Neurol Neurosurg Psychiatry, 85, 668674. doi:10.1136/jnnp-2013-306439.Google Scholar
Drui, G, Carnicella, S, Carcenac, C, et al. (2014). Loss of dopaminergic nigrostriatal neurons accounts for the motivational and affective deficits in Parkinson’s disease. Mol Psychiatry, 19, 358367. doi:10.1038/mp.2013.3.Google Scholar
Ecker, D, Unrath, A, Kassubek, J, Sabolek, M (2009). Dopamine agonists and their risk to induce psychotic episodes in Parkinson’s disease: a case-control study. BMC Neurol, 9, 23. doi:10.1186/1471-2377-9-23.Google Scholar
Emre, M, Tsolaki, M, Bonucelli, U, et al. (2010). Memantine for patients with Parkinson’s disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trial. Lancet Neurol, 9(10), 969977.Google Scholar
Engedal, K, Davis, B, Richarz, U, et al. (2012). Two galantamine titration regimens in patients switched from donepezil. Acta Neurol Scand, 126, 3744. doi:10.1111/j.1600-0404.2011.01594.x.Google Scholar
Fox, C, Livingston, G, Maidment, ID, et al. (2011). The impact of anticholinergic burden in Alzheimer’s dementia – the laser-AD study. Age Ageing, 40, 730735.Google Scholar
Ghaleiha, A, Ghyasvand, M, Mohammadi, MR, et al. (2014). Galantamine efficacy and tolerability as an augmentative therapy in autistic children: a randomized, double-blind, placebo-controlled trial. J Psychopharmacol, 28, 677685.Google Scholar
Glasgow, NG, Povysheva, NV, Azofeifa, AM, Johnson, JW (2017). Memantine and ketamine differentially alter NMDA receptor desensitization. J Neurosci, 37, 96869704.Google Scholar
Gobburu, JVS, Tammara, V, Lesko, L, et al. (2001). Pharmacokinetic-pharmacodynamic modeling of rivastigmine, a cholinesterase inhibitor, in patients with Alzheimer’s disease. J Clin Pharmacol, 41, 10821090.Google Scholar
Green, AR, Oh, E, Hilson, L, Tian, J, Boyd, CM (2016). Anticholinergic burden in older adults with mild cognitive impairment. J Am Geriatr Soc, 64, e313e314.Google Scholar
Grossberg, GT, Manes, F, Allegri, RF, et al. (2013). The safety, tolerability, and efficacy of once-daily memantine (28 mg): a multinational, randomized, double-blind, placebo-controlled trial in patients with moderate-to-severe Alzheimer’s disease taking cholinesterase inhibitors. CNS Drugs, 27, 469478.Google Scholar
Haddad, PM, Benbow, SM (1993a). Sexual problems associated with dementia: Part 1. Problems and their consequences. Int J Geriatr Psychiatry, 8(7), 547551.Google Scholar
Haddad, PM, Benbow, SM (1993b). Sexual problems associated with dementia: Part 2. Aetiology, assessment and treatment. Int J Geriatr Psychiatry, 8(8), 631637.Google Scholar
Herrmann, N, Mamdani, M, Lanctôt, KL (2004). Atypical antipsychotics and risk of cerebrovascular accidents. Am J Psychiatry, 161, 11131115. doi:10.1176/appi.ajp.161.6.1113.Google Scholar
Hilmas, CJ, Poole, MJ, Finneran, K, Clark, MG, Williams, PT (2009). Galantamine is a novel post-exposure therapeutic against lethal VX challenge. Toxicol Appl Pharmacol, 240, 166173.Google Scholar
Hinkle, JT, Perepezko, K, Bakker, C, et al. (2018). Onset and remission of psychosis in Parkinson’s disease: pharmacologic and motoric markers. Mov Disord Clin Pract, 5, 3138. doi:10.1002/mdc3.12550.Google Scholar
Holroyd, S, Currie, L, Wooten, GF (2001). Prospective study of hallucinations and delusions in Parkinson’s disease. J Neurol Neurosurg Psychiatry, 70, 734738. doi:10.1136/jnnp.70.6.734.Google Scholar
Hong, JM, Shin, DH, Lim, TS, Lee, JS, Huh, K (2012). Galantamine administration in chronic post-stroke aphasia. J Neurol Neurosurg Psychiatry, 83, 675680. doi:10.1136/jnnp-2012-302268.Google Scholar
Howard, R, McShane, R, Lindesay, J, et al. (2012). Donepezil and memantine for moderate-to-severe Alzheimer’s disease. N Engl J Med, 366, 893903.Google Scholar
Howard, R, McShane, R, Lindesay, J, et al. (2015). Nursing home placement in the Donepezil and Memantine in Moderate to Severe Alzheimer’s Disease (DOMINO-AD) trial: secondary and post-hoc analyses. Lancet Neurol, 14, 11711181.Google Scholar
Huang, F, Lasseter, AC, Janssens, L, et al. (2002). Pharmacokinetic and safety assessments of galantamine and risperidone after the two drugs are administered alone and together. J Clin Pharmacol, 42, 13411351.Google Scholar
Irwin, DJ, Grossman, M, Weintraub, D, et al. (2017). Neuropathological and genetic correlates of survival and dementia onset in synucleinopathies: a retrospective analysis. Lancet Neurol, 16, 5565.Google Scholar
Jang, YK, Lyoo, CH, Park, S, et al. (2018). Head to head comparison of [18F] AV-1451 and [18F] THK5351 for tau imaging in Alzheimer’s disease and frontotemporal dementia. Eur J Nucl Med Mol Imaging, 45, 432442.Google Scholar
Jann, MW, Shirley, KL, Small, GW (2002). Clinical pharmacokinetics and pharmacodynamics of cholinesterase inhibitors. Clin Pharmacokinet, 41, 719739.Google Scholar
Jiang, J, Jiang, H (2015). Efficacy and adverse effects of memantine treatment for Alzheimer’s disease from randomized controlled trials. Neurol Sci, 36, 16331641.Google Scholar
Johansson, I, Nordberg, A (1993). Pharmacokinetic studies of cholinesterase inhibitors. Acta Neurol Scand Suppl, 149, 2225.Google Scholar
Kavirajan, H, Schneider, LS (2007). Efficacy and adverse effects of cholinesterase inhibitors and memantine in vascular dementia: a meta-analysis of randomised controlled trials. Lancet Neurol, 6, 782792. doi:10.1016/S1474-4422(07)70195-3.Google Scholar
Kishi, T, Matsunaga, S, Iwata, N (2015). Memantine for the treatment of frontotemporal dementia: a meta-analysis. Neuropsychiatr Dis Treat, 11, 28832885.Google Scholar
Kobayashi, H, Ohnishi, T, Nakagawa, R, Yoshizawa, K (2016). The comparative efficacy and safety of cholinesterase inhibitors in patients with mild-to-moderate Alzheimer’s disease: a Bayesian network meta-analysis. Int J Geriatr Psychiatry, 31, 892904.Google Scholar
Larsson, V, Aarsland, D, Ballard, C, Minthon, L, Londos, E (2010). The effect of memantine on sleep behaviour in dementia with Lewy bodies and Parkinson’s disease dementia. Int J Geriatr Psychiatry, 25, 10301038. doi:10.1002/gps.2506.Google Scholar
Lefèvre, G, Callegari, F, Gsteiger, S, Xiong, Y (2016). Effects of renal impairment on steady-state plasma concentrations of rivastigmine: a population pharmacokinetic analysis of capsule and patch formulations in patients with Alzheimer’s disease. Drugs Aging, 33, 725736.Google Scholar
Liu, AKL, Chang, RCC, Pearce, RKB, Gentleman, SM (2015). Nucleus basalis of Meynert revisited: anatomy, history and differential involvement in Alzheimer’s and Parkinson’s disease. Acta Neuropathol, 129, 527540. doi:10.1007/s00401-015-1392-5.Google Scholar
Livingston, G, Kelly, J, Lewis-Holmes, E, et al. (2014). A systematic review of the clinical effectiveness and cost-effectiveness of sensory, psychological and behavioural interventions for managing agitation in older adults with dementia. Health Technol Assess, 18, 1226, v–vi.Google Scholar
López-Valdés, HE, Clarkson, AN, Ao, Y, et al. (2014). Memantine enhances recovery from stroke. Stroke, 45, 20932100. doi:10.1161/STROKEAHA.113.004476.Google Scholar
Massoud, F, Desmarais, JE, Gauthier, S (2011). Switching cholinesterase inhibitors in older adults with dementia. Int Psychogeriatr, 23, 372378. doi:10.1017/S1041610210001985.Google Scholar
Matsunaga, S, Kishi, T, Iwata, N (2015a). Memantine monotherapy for Alzheimer’s disease: a systematic review and meta-analysis. PLoS One, 10(4), e0123289. doi:10.1371/journal.pone.0123289.Google Scholar
Matsunaga, S, Kishi, T, Iwata, N (2015b). Combination therapy with cholinesterase inhibitors and memantine for Alzheimer’s disease: a systematic review and meta-analysis. Int J Neuropsychopharmacol, 18, pyu115. doi:10.1093/ijnp/pyu115.Google Scholar
Meltzer, HY, Mills, R, Revell, S, et al. (2010). Pimavanserin, a serotonin 2 A receptor inverse agonist, for the treatment of Parkinson’s disease psychosis. Neuropsychopharmacology, 35, 881892.Google Scholar
National Institute for Health and Care Excellence (NICE) (2018a). Decision aid: antipsychotic medicines for treating agitation, aggression and distress in people living with dementia. London: National Institute for Health and Care Excellence. Available at: www.nice.org.uk/guidance/ng97/resources/antipsychotic-medicines-for-treating-agitation-aggression-and-distress-in-people-living-with-dementia-patient-decision-aid-pdf-4852697005 (last accessed 26.1.19).Google Scholar
National Institute for Health and Care Excellence (NICE) (2018b). Dementia: assessment, management and support for people living with dementia and their carers. NICE guideline [NG97]. London: National Institute for Health and Care Excellence.Google Scholar
Nelson, JC, Devanand, DP (2011). A systematic review and meta-analysis of placebo-controlled antidepressant studies in people with depression and dementia. J Am Geriatr Soc, 59, 577585. doi:10.1111/j.1532-5415.2011.03355.x.Google Scholar
Noetzli, M, Guidi, M, Ebbing, K, et al. (2013). Population pharmacokinetic study of memantine: effects of clinical and genetic factors. Clin Pharmacokinet, 52, 211223.Google Scholar
Ohnishi, A, Mihara, M, Kamakura, H, et al. (1993). Comparison of the pharmacokinetics of E2020, a new compound for Alzheimer’s disease, in healthy young and elderly subjects. J Clin Pharmacol, 33, 10861091.Google Scholar
Ohta, Y, Darwish, M, Hishikawa, N, et al. (2017). Therapeutic effects of drug switching between acetylcholinesterase inhibitors in patients with Alzheimer’s disease. Geriatr Gerontol Int, 17, 18431848. doi:10.1111/ggi.12971.Google Scholar
Pagonabarraga, J, Kulisevsky, J, Strafella, AP, Krack, P (2015). Apathy in Parkinson’s disease: clinical features, neural substrates, diagnosis, and treatment. Lancet Neurology, 14, 518531. doi:10.1016/S1474-4422(15)00019-8.Google Scholar
Pasina, L, Djade, CD, Lucca, U, et al. (2013). Association of anticholinergic burden with cognitive and functional status in a cohort of hospitalized elderly: comparison of the anticholinergic cognitive burden scale and anticholinergic risk scale: results from the REPOSI study. Drugs Aging, 30, 103112.Google Scholar
Pereira, EFR, Aracava, Y, Alkondon, M, et al. (2010). Molecular and cellular actions of galantamine: clinical implications for treatment of organophosphorus poisoning. J Mol Neurosci, 40, 196203.Google Scholar
Periclou, A, Ventura, D, Rao, N, Abramowitz, W (2006). Pharmacokinetic study of memantine in healthy and renally impaired subjects. Clin Pharmacol Ther, 79, 134143.Google Scholar
Perry, EK, Curtis, M, Dick, DJ, et al. (1985). Cholinergic correlates of cognitive impairment in Parkinson’s disease: comparisons with Alzheimer’s disease. J Neurol Neurosurg Psychiatry, 48, 413421.Google Scholar
Piotrovsky, V, Van Peer, A, Van Osselaer, N, Armstrong, M, Aerssens, J (2003). Galantamine population pharmacokinetics in patients with Alzheimer’s disease: modeling and simulations. J Clin Pharmacol, 43, 514523.Google Scholar
Polinsky, RJ (1998). Clinical pharmacology of rivastigmine: a new-generation acetylcholinesterase inhibitor for the treatment of Alzheimer’s disease. Clin Ther, 20, 634647.Google Scholar
Prince, M, Wimo, A, Guerchet, M, et al. (2015). World Alzheimer Report 2015 – The Global Impact of Dementia: An Analysis of Prevalence, Incidence, Cost and Trends. London: Alzheimer’s Disease International. Available at: www.alz.co.uk/research/WorldAlzheimerReport2015.pdf (last accessed 10.8.19).Google Scholar
Risacher, SL, McDonald, BC, Tallman, EF, et al. (2016). Association between anticholinergic medication use and cognition, brain metabolism, and brain atrophy in cognitively normal older adults. JAMA Neurol, 73, 721732.Google Scholar
Ruxton, K, Woodman, RJ, Mangoni, AA (2015). Drugs with anticholinergic effects and cognitive impairment, falls and all-cause mortality in older adults: a systematic review and meta-analysis. Br J Clin Pharmacol, 80, 209220.Google Scholar
Sabbagh, M, Hans, S, Kim, S, et al. (2016). Clinical recommendations for the use of donepezil 23 mg in moderate-to-severe Alzheimer’s disease in the Asia-Pacific region. Dement Geriatr Cogn Dis Extra, 6, 382395.Google Scholar
Salahudeen, MS, Duffull, SB, Nishtala, PS (2015). Anticholinergic burden quantified by anticholinergic risk scales and adverse outcomes in older people: a systematic review. BMC Geriatr, 15, 31.Google Scholar
Sasaki, S, Horie, Y (2014). The effects of an uninterrupted switch from donepezil to galantamine without dose titration on behavioral and psychological symptoms of dementia in Alzheimer’s disease. Dement Geriatr Cogn Dis Extra, 4, 131139. doi:10.1159/000362871.Google Scholar
Schmidt, R, Hofer, E, Bouwman, FH, et al. (2015). EFNS-ENS/EAN Guideline on concomitant use of cholinesterase inhibitors and memantine in moderate to severe Alzheimer’s disease. Eur J Neurol, 22, 889898.Google Scholar
Schneider-Thoma, J, Efthimiou, O, Huhn, M, et al. (2018). Second-generation antipsychotic drugs and short-term mortality: a systematic review and meta-analysis of placebo-controlled randomised controlled trials. Lancet Psychiatry, 5, 653663.Google Scholar
Serfaty, M, Kennell-Webb, S, Warner, J, Blizard, R, Raven, P (2002). Double blind randomised placebo controlled trial of low dose melatonin for sleep disorders in dementia. Int J Geriatr Psychiatry, 17, 11201127.Google Scholar
Sharp, SI, Francis, PT, Elliot, MSJ, et al. (2009). Choline acetyltransferase activity in vascular dementia and stroke. Dement Geriatr Cogn Disord, 28, 233238. doi:10.1159/000239235.Google Scholar
Sullivan, SC, Richards, KC (2004). Predictors or circadian sleep-wake rhythm maintenance in elders with dementia. Aging Ment Health, 8, 143152. doi:10.1080/13607860410001649608.Google Scholar
Tariot, PN, Farlow, MR, Grossberg, GT, et al. (2004). Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil. JAMA, 291, 317324.Google Scholar
Thobois, S, Lhommée, E, Klinger, H, et al. (2013). Parkinsonian apathy responds to dopaminergic stimulation of D2/D3 receptors with piribedil. Brain, 136, 15681577.Google Scholar
Trotman, M, Vermehren, P, Gibson, CL, Fern, R (2015). The dichotomy of memantine treatment for ischemic stroke: dose-dependent protective and detrimental effects. J Cereb Blood Flow Metab, 35, 230239. doi:10.1038/jcbfm.2014.188.Google Scholar
van Gool, WA, van de Beek, D, Eikelenboom, P (2010). Systemic infection and delirium: when cytokines and acetylcholine collide. Lancet, 375(9716), 773775. doi:10.1016/S0140-6736(09)61158-2.Google Scholar
Van Leeuwen, E, Petrovic, M, Azermai, M, et al. (2018). Withdrawal versus continuation of long-term antipsychotic drug use for behavioural and psychological symptoms in older people with dementia. Cochrane Database Syst Rev, (3), CD007726.Google Scholar
Vingtdeux, V, Dreses-Werringloer, U, Zhao, H, Davies, P, Marambaud, P (2008). Therapeutic potential of resveratrol in Alzheimer’s disease. BMC Neurosci, 9, S6.Google Scholar
Wang, J, Yu, JT, Wang, HF, et al. (2015). Pharmacological treatment of neuropsychiatric symptoms in Alzheimer’s disease: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry, 86, 101109.Google Scholar
Weintraub, D, Chiang, C, Kim, HM, et al. (2016). Association of antipsychotic use with mortality risk in patients with Parkinson disease. JAMA Neurol, 73, 535541. doi:10.1001/jamaneurol.2016.0031.Google Scholar
Wesnes, K, Aarsland, D, Ballard, C, Londos, E (2014). Memantine improves attention and episodic memory in Parkinson’s disease dementia and dementia with Lewy bodies. Int J Geriatr Psychiatry, 30(1), 4654.Google Scholar
Zhang, L-J, Sha, X-Y (2007). Pharmacokinetics and bioequivalence studies of galantamine hydrobromide dispersible tablet in healthy male Chinese volunteers. Drug Dev Ind Pharm, 33, 335340.Google Scholar
Zhao, Q, Janssens, L, Verhaeghe, T, Brashear, HR, Truyen, L (2005). Pharmacokinetics of extended-release and immediate-release formulations of galantamine at steady state in healthy volunteers. Curr Med Res Opin, 21, 15471554.Google Scholar
Zhao, QF, Tan, L, Wang, HF, et al. (2016). The prevalence of neuropsychiatric symptoms in Alzheimer’s disease: systematic review and meta-analysis. J Affect Disord, 190, 264271.Google Scholar

References

Agabio, R, Sinclair, J, Addolorato, G, et al. (2018). Baclofen for the treatment of alcohol use disorder: the Cagliari Statement. Lancet Psychiatry, 5, 957960.Google Scholar
Amato, L, Minozzi, S, Davoli, M (2011). Efficacy and safety of pharmacological interventions for the treatment of the Alcohol Withdrawal Syndrome. Cochrane Database Syst Rev, (6), CD008537.Google Scholar
American Psychiatric Association (2013). Diagnostic and Statistical Manual of Mental Disorders, 5th ed. Arlington, VA: American Psychiatric Association.Google Scholar
Anthenelli, RM, Benowitz, NL, West, R, et al. (2016). Neuropsychiatric safety and efficacy of varenicline, bupropion, and nicotine patch in smokers with and without psychiatric disorders (EAGLES): a double-blind, randomised, placebo-controlled clinical trial. Lancet, 387, 25072520.Google Scholar
Bakker, A, Streel, E (2016). Benzodiazepine maintenance in opiate substitution treatment: good or bad? A retrospective primary care case-note review. J Psychopharmacol, 31, 6266.Google Scholar
Beck, T, Haasen, C, Verthein, U, et al. (2014). Maintenance treatment for opioid dependence with slow-release oral morphine: a randomized cross-over, non-inferiority study versus methadone. Addiction, 109, 617626.Google Scholar
Bordia, T, Hrachova, M, Chin, M, McIntosh, JM, Quik, M (2012). Varenicline is a potent partial agonist at α6β2* nicotinic acetylcholine receptors in rat and monkey striatum. J Pharmacol Exp Ther, 342, 327334.Google Scholar
Burns, L, Gisev, N, Larney, S, et al. (2015). A longitudinal comparison of retention in buprenorphine and methadone treatment for opioid dependence in New South Wales, Australia. Addiction, 110, 646655.Google Scholar
Cahill, K, Lindson-Hawley, N, Thomas, KH, Fanshawe, TR, Lancaster, T (2016). Nicotine receptor partial agonists for smoking cessation. Cochrane Database Syst Rev, (5), CD006103.Google Scholar
Chick, J (1999). Safety issues concerning the use of disulfiram in treating alcohol dependence. Drug Saf, 20, 427435.Google Scholar
Chick, J, Nutt, DJ (2012). Substitution therapy for alcoholism: time for a reappraisal? J Psychopharmacol, 26, 205212.Google Scholar
Chick, J, Gough, K, Falkowski, W, et al. (1992). Disulfiram treatment of alcoholism. Br J Psychiatry, 161, 8489.Google Scholar
Clinical Guidelines on Drug Misuse and Dependence Update 2017 Independent Expert Working Group (2017). Drug misuse and dependence: UK guidelines on clinical management. London: Department of Health.Google Scholar
Coe, JW, Brooks, PR, Vetelino, MG, et al. (2005). Varenicline: an α4β2 nicotinic receptor partial agonist for smoking cessation. J Med Chem, 48, 34743477.Google Scholar
Colasanti, A, Rabiner, E, Lingford-Hughes, A, Nutt, D (2011). Opioids and anxiety. J Psychopharmacol, 25, 14151433.Google Scholar
Coleman, T, Chamberlain, C, Davey, M-A, Cooper, SE, Leonardi-Bee, J (2015). Pharmacological interventions for promoting smoking cessation during pregnancy. Cochrane Database Syst Rev, (12), CD010078.Google Scholar
Darke, S, Ross, J, Mills, K, et al. (2010). Benzodiazepine use among heroin users: baseline use, current use and clinical outcome. Drug Alcohol Rev, 29, 250255.Google Scholar
Darker, CD, Sweeney, BP, Barry, JM, Farrell, MF, Donnelly-Swift, E (2015). Psychosocial interventions for benzodiazepine harmful use, abuse or dependence. Cochrane Database Syst Rev, (5), CD009652.Google Scholar
de Beaurepaire, R, Sinclair, JMA, Heydtmann, M, et al. (2018). The use of baclofen as a treatment for alcohol use disorder: a clinical practice perspective. Front Psychiatry, 9, 708.Google Scholar
Degenhardt, L, Charlson, F, Mathers, B, et al. (2014a). The global epidemiology and burden of opioid dependence: results from the global burden of disease 2010 study. Addiction, 109, 13201333.Google Scholar
Degenhardt, L, Larney, S, Randall, D, Burns, L, Hall, W (2014b). Causes of death in a cohort treated for opioid dependence between 1985 and 2005. Addiction, 109, 9099.Google Scholar
Di Ciano, P, Guranda, M, Lagzdins, D, et al. (2016). Varenicline-induced elevation of dopamine in smokers: a preliminary [11C]-(+)-PHNO PET study. Neuropsychopharmacology, 41, 15131520.Google Scholar
Donoghue, K, Elzerbi, C, Saunders, R, et al. (2015). The efficacy of acamprosate and naltrexone in the treatment of alcohol dependence, Europe versus the rest of the world: a meta‐analysis. Addiction, 110, 920930.Google Scholar
Ebbert, JO, Hughes, JR, West, RJ, et al. (2015). Effect of varenicline on smoking cessation through smoking reduction: a randomized clinical trial. JAMA, 313, 687694.Google Scholar
Elholm, B, Larsen, K, Hornnes, N, Zierau, F, Becker, U (2011). Alcohol withdrawal syndrome: symptom-triggered versus fixed-schedule treatment in an outpatient setting. Alcohol Alcohol, 46, 318323.Google Scholar
European Medicines Agency (2017). Alcover (granules in sachet) and associated names. London: European Medicines Agency.Google Scholar
Garcia-Borreguero, D, Bronisch, T, Apelt, S, Yassouridis, A, Emrich, HM (1991). Treatment of benzodiazepine withdrawal symptoms with carbamazepine. Eur Arch Psychiatry Clin Neurosci, 241, 145150.Google Scholar
Goodwin, G, Haddad, P, Ferrier, I, et al. (2016). Evidence-based guidelines for treating bipolar disorder: revised third edition recommendations from the British Association for Psychopharmacology. J Psychopharmacol, 30, 495553.Google Scholar
Gossop, M (1990). The development of a short opiate withdrawal scale (SOWS). Addict Behav, 15, 487490.Google Scholar
Gowing, L, Farrell, M, Ali, R, White, JM (2016). Alpha2-adrenergic agonists for the management of opioid withdrawal. Cochrane Database Syst Rev, (5), CD002024.Google Scholar
Haber, P, Lintzeris, N, Proude, E, Lopatko, O (2009). Guidelines for the Treatment of Alcohol Problems. Canberra: Commonwealth of Australia.Google Scholar
Hajak, G, Müller, WE, Wittchen, HU, Pittrow, D, Kirch, W (2003). Abuse and dependence potential for the non-benzodiazepine hypnotics zolpidem and zopiclone: a review of case reports and epidemiological data. Addiction, 98, 13711378.Google Scholar
Harper, CG, Giles, M, Finlay-Jones, R (1986). Clinical signs in the Wernicke-Korsakoff complex: a retrospective analysis of 131 cases diagnosed at necropsy. J Neurol Neurosurg Psychiatry, 49, 341345.Google Scholar
Hartmann-Boyce, J, McRobbie, H, Bullen, C, et al. (2016). Electronic cigarettes for smoking cessation. Cochrane Database Syst Rev, (9), CD010216.Google Scholar
Hartmann-Boyce, J, Chepkin, SC, Ye, W, Bullen, C, Lancaster, T (2018). Nicotine replacement therapy versus control for smoking cessation. Cochrane Database Syst Rev, (5), CD000146.Google Scholar
Hasin, DS, Stinson, FS, Ogburn, E, Grant, BF (2007). Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States: results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry, 64, 830842.Google Scholar
Hay, G, Rael dos Santos, A, Swithenbank, Z (2017). Estimates of the Prevalence of Opiate Use and/or Crack Cocaine Use, 2014/15: Sweep 11 Report. Public Health England. Liverpool: Public Health Institute, John Moores University.Google Scholar
Hser, Y-I, Evans, E, Huang, D, et al. (2016). Long-term outcomes after randomization to buprenorphine/naloxone versus methadone in a multi-site trial. Addiction, 111, 695705.Google Scholar
Hughes, JR, Stead, LF, Hartmann-Boyce, J, Cahill, K, Lancaster, T (2014). Antidepressants for smoking cessation. Cochrane Database Syst Rev, (1), CD000031.Google Scholar
Jones, HE, Kaltenbach, K, Heil, SH, et al. (2010). Neonatal abstinence syndrome after methadone or buprenorphine exposure. New Engl J Med, 363, 23202331.Google Scholar
Kalk, NJ, Lingford-Hughes, AR (2014). The clinical pharmacology of acamprosate. Br J Clin Pharmacol, 77, 315323.Google Scholar
Koob, GF, Le Moal, M (2006). Nicotine. In Koob, GF, Le Moal, M, eds., Neurobiology of Addiction. Amsterdam: Elsevier.Google Scholar
Kosten, TR, George, TP (2002). The neurobiology of opioid dependence: implications for treatment. Sci Pract Perspect, 1, 1320.Google Scholar
Kotz, D, Viechtbauer, W, Simpson, C, et al. (2015). Cardiovascular and neuropsychiatric risks of varenicline: a retrospective cohort study. Lancet Respir Med, 3, 761768.Google Scholar
Larney, S, Gowing, L, Mattick, RP, et al. (2014). A systematic review and meta-analysis of naltrexone implants for the treatment of opioid dependence. Drug Alcohol Rev, 33, 115128.Google Scholar
Lasser, K, Boyd, J, Woolhandler, S, et al. (2000). Smoking and mental illness: a population-based prevalence study. JAMA, 284, 26062610.Google Scholar
Law, FD, Diaper, AM, Melichar, JK, et al. (2017). Buprenorphine/naloxone versus methadone and lofexidine in community stabilisation and detoxification: a randomised controlled trial of low dose short-term opiate-dependent individuals. J Psychopharamcol, 31, 10461055.Google Scholar
Lejoyeux, M, Lehert, P (2011). Alcohol-use disorders and depression: results from individual patient data meta-analysis of the acamprosate-controlled studies. Alcohol Alcohol, 46, 6167.Google Scholar
Liebrenz, M, Boesch, L, Stohler, R, Caflisch, C (2010). Agonist substitution – a treatment alternative for high-dose benzodiazepine-dependent patients? Addiction, 105, 18701874.Google Scholar
Lingford-Hughes, AR, Welch, S, Peters, L, Nutt, DJ (2012). BAP updated guidelines: evidence-based guidelines for the pharmacological management of substance abuse, harmful use, addiction and comorbidity: recommendations from BAP. J Psychopharmacol, 26, 899952.Google Scholar
Lishman, W (1998). Organic Psychiatry: The Psychological Consequences of Cerebral Disorder. Oxford: Blackwell Science, pp. 315323.Google Scholar
Mann, K, Torup, L, Sørensen, P, et al. (2016). Nalmefene for the management of alcohol dependence: review on its pharmacology, mechanism of action and meta-analysis on its clinical efficacy. Eur Neuropsychopharmacol, 26, 19411949.Google Scholar
Marsch, LA, Moore, SK, Borodovsky, JT, et al. (2016). A randomized controlled trial of buprenorphine taper duration among opioid-dependent adolescents and young adults. Addiction, 111, 14061415.Google Scholar
Mattick, RP, Breen, C, Kimber, J, Davoli, M (2009). Methadone maintenance therapy versus no opioid replacement therapy for opioid dependence. Cochrane Database Syst Rev, (3), CD002209.Google Scholar
Mattick, RP, Breen, C, Kimber, J, Davoli, M (2014). Buprenorphine maintenance versus placebo or methadone maintenance for opioid dependence. Cochrane Database of Systematic Reviews, (2), CD002207.Google Scholar
McNeill, A, Brose, LS, Calder, R, Bauld, L, Robson, D (2018). Evidence review of e-cigarettes and heated tobacco products. A report commissioned by Public Health England. London: Public Health England.Google Scholar
MHRA (2006). Current Problems in Pharmacovigilance, Vol. 31. London: Medicines and Healthcare products Regulatory Agency.Google Scholar
Minozzi, S, Amato, L, Vecchi, S, Davoli, M (2010). Anticonvulsants for alcohol withdrawal. Cochrane Database Syst Rev, (3), CD005064.Google Scholar
Minozzi, S, Amato, L, Vecchi, S, et al. (2011). Oral naltrexone maintenance treatment for opioid dependence. Cochrane Database Syst Rev, (4), CD001333.Google Scholar
Montgomery, AJ, Lingford‐Hughes, AR, Egerton, A, Nutt, DJ, Grasby, PM (2007). The effect of nicotine on striatal dopamine release in man: a [11C] raclopride PET study. Synapse, 61, 637645.Google Scholar
National Institute for Health and Care Excellence (NICE) (2007a). Methadone and buprenorphine for the management of opioid dependence. Technology appraisal guidance [TA114]. Manchester: National Institute for Health and Care Excellence.Google Scholar
National Institute for Health and Care Excellence (NICE) (2007b). Drug misuse: opiate detoxification. Clinical guideline [CG52]. Manchester: National Institute for Health and Care Excellence.Google Scholar
National Institute for Health and Care Excellence (NICE) (2007c). Naltrexone for the management of opioid dependence. Technology appraisal guidance [TA115]. Manchester: National Institute for Health and Care Excellence.Google Scholar
National Institute for Health and Care Excellence (NICE) (2010a). Alcohol-use disorders: diagnosis and management of physical complications. Clinical guideline [CG100]. Manchester: National Institute for Health and Care Excellence.Google Scholar
National Institute for Health and Care Excellence (NICE) (2010b). Smoking: stopping in pregnancy and after childbirth. Public health guideline [PH26]. Manchester: National Institute for Health and Care Excellence.Google Scholar
National Institute for Health and Care Excellence (NICE) (2011). Alcohol-use disorders: diagnosis, assessment and management of harmful drinking and alcohol dependence. Clinical guideline [CG115]. Manchester: National Institute for Health and Care Excellence.Google Scholar
Nesvåg, R, Knudsen, GP, Bakken, IJ, et al. (2015). Substance use disorders in schizophrenia, bipolar disorder, and depressive illness: a registry-based study. Soc Psychiatry Psychiatr Epidemiol, 50, 12671276.Google Scholar
Nielsen, S, Dietze, P, Lee, N, Dunlop, A, Taylor, D (2007). Concurrent buprenorphine and benzodiazepines use and self-reported opioid toxicity in opioid substitution treatment. Addiction, 102, 616622.Google Scholar
Nolan, S, Dias Lima, V, Fairbairn, N, et al. (2014). The impact of methadone maintenance therapy on hepatitis C incidence among illicit drug users. Addiction, 109, 20532059.Google Scholar
Nosyk, B, Sun, H, Evans, E, et al. (2012). Defining dosing pattern characteristics of successful tapers following methadone maintenance treatment: results from a population-based retrospective cohort study. Addiction, 107, 16211629.Google Scholar
Novel Psychoactive Treatment UK Network (NEPTUNE) (2015). Guidance on the management of acute and chronic harms of club drugs and novel psychoactive substances. (online). London. (last accessed 19.2.18).Google Scholar
Nutt, DJ, Lingford-Hughes, A, Chick, J (2012). Through a glass darkly: can we improve clarity about mechanism and aims of medications in drug and alcohol treatments? J Psychopharmacol, 26, 199204.Google Scholar
Nutt, DJ, Lingford-Hughes, A, Erritzoe, D, Stokes, P (2015). The dopamine theory of addiction: 40 years of highs and lows. Nat Rev Neurosci, 16, 305312.Google Scholar
Office for National Statistics (2017a). Deaths related to drug poisoning in England and Wales: 2016 registrations. Statistical Bulletin. London: Office for National Statistics.Google Scholar
Office for National Statistics (2017b). Drug misuse: findings from the 2016/17 CSEW. London: Home Office.Google Scholar
Pathan, H, Williams, J (2012). Basic opioid pharmacology: an update. Br J Pain, 6, 1116.Google Scholar
Petrakis, IL, Poling, J, Levinson, C, et al. (2005). Naltrexone and disulfiram in patients with alcohol dependence and comorbid psychiatric disorders. Biol Psychiatry, 57, 11281137.Google Scholar
Pierce, M, Bird, SM, Hickman, M, et al. (2016). Impact of treatment for opioid dependence on fatal drug-related poisoning: a national cohort study in England. Addiction, 111, 298308.Google Scholar
Public Health England (2016). Shooting up: infections among people who injected drugs in the UK, 2015. An update: November 2016. London: Public Health England.Google Scholar
Public Health England (2017a). Take-home naloxone for opioid overdose in people who use drugs. London: Public Health England.Google Scholar
Public Health England (2017b). Adult substance misuse statistics from the National Drug Treatment Monitoring System (NDTMS). London: Public Health England.Google Scholar
Public Health England (2018). Adult substance misuse statistics from the National Drug Treatment Monitoring System (NDTMS). London: Public Health England.Google Scholar
Quelch, DR, Mick, I, McGonigle, J, et al. (2017). Nalmefene reduces reward anticipation in alcohol dependence: an experimental functional magnetic resonance imaging study. Biol Psychiatry, 81, 941948.Google Scholar
Rehm, J, Shield, K, Gmel, G, Rehm, M, Frick, U (2013). Modeling the impact of alcohol dependence on mortality burden and the effect of available treatment interventions in the European Union. Eur Neuropsychopharmacol, 23, 8997.Google Scholar
Reitsma, M (2017). Smoking prevalence and attributable disease burden in 195 countries and territories, 1990–2015: a systematic analysis from the Global Burden of Disease Study 2015. Lancet, 389, 18851906.Google Scholar
Robertson, JR, Raab, GM, Bruce, M, et al. (2006). Addressing the efficacy of dihydrocodeine versus methadone as an alternative maintenance treatment for opiate dependence: a randomized controlled trial. Addiction, 101, 17521759.Google Scholar
Rösner, S, Hackl‐Herrwerth, A, Leucht, S, et al. (2010). Opioid antagonists for alcohol dependence. Cochrane Database Syst Rev, (12), CD001867.Google Scholar
Ross, J, Ross, J, Teesson, M, et al. (2005). The characteristics of heroin users entering treatment: findings from the Australian treatment outcome study (ATOS). Drug Alcohol Rev, 24, 411418.Google Scholar
Saunders, JB, Aasland, OG, Amundsen, A, Grant, M (1993). Alcohol consumption and related problems among primary health care patients: WHO collaborative project on early detection of persons with harmful alcohol consumption – I. Addiction, 88, 349362.Google Scholar
Sechi, G, Serra, A (2007). Wernicke’s encephalopathy: new clinical settings and recent advances in diagnosis and management. Lancet Neurol, 6, 442455.Google Scholar
Senbanjo, R, Wolff, K, Marshall, J (2007). Excessive alcohol consumption is associated with reduced quality of life among methadone patients. Addiction, 102, 257263.Google Scholar
Sinclair, JMA, Latifi, AH, Latifi, AW (2008). Co-morbid substance misuse in psychiatric inpatients: prevalence and association with length of inpatient stay. J Psychopharmacol, 22, 9298.Google Scholar
Sinclair, JM, Chambers, SE, Shiles, CJ, Baldwin, DS (2016). Safety and tolerability of pharmacological treatment of alcohol dependence: comprehensive review of evidence. Drug Saf, 39, 627645.Google Scholar
Sordo, L, Barrio, G, Bravo, MJ, et al. (2017). Mortality risk during and after opioid substitution treatment: systematic review and meta-analysis of cohort studies. BMJ, 357, j1550.Google Scholar
Stapleton, J, West, R, Hajek, P, et al. (2013). Randomized trial of nicotine replacement therapy (NRT), bupropion and NRT plus bupropion for smoking cessation: effectiveness in clinical practice. Addiction, 108, 21932201.Google Scholar
Sterling, LH, Windle, SB, Filion, KB, Touma, L, Eisenberg, MJ (2016). Varenicline and adverse cardiovascular events: a systematic review and meta‐analysis of randomized controlled trials. J Am Heart Assoc, 5, e002849.Google Scholar
Stockwell, T, Murphy, D, Hodgson, R (1983). The severity of alcohol dependence questionnaire: its use, reliability and validity. Br J Addict, 78, 145155.Google Scholar
Strang, J, Groshkova, T, Uchtenhagen, A, et al. (2015). Heroin on trial: systematic review and meta-analysis of randomised trials of diamorphine-prescribing as treatment for refractory heroin addiction. Br J Psychiatry, 207, 514.Google Scholar
Sullivan, JT, Sykora, K, Schneiderman, J, Naranjo, CA, Sellers, EM (1989). Assessment of alcohol withdrawal: the revised Clinical Institute Withdrawal Assessment for Alcohol scale (CIWA-Ar). Br J Addict, 84, 1353–1357.Google Scholar
Sun, EC, Dixit, A, Humphreys, K, et al. (2017). Association between concurrent use of prescription opioids and benzodiazepines and overdose: retrospective analysis. BMJ, 356, j760.Google Scholar
Sweizer, E, Rickets, K, Case, WG, Greenblatt, DJ (1991). Carbamazepine treatment in patients discontinuing long-term benzodiazepine therapy. Effects on withdrawal severity and outcome. Arch Gen Psychiatry, 48, 448452.Google Scholar
Swift, RM (2013). Naltrexone and nalmefene: any meaningful difference? Biol Psychiatry, 73, 700701.Google Scholar
Toftdahl, NG, Nordentoft, M, Hjorthøj, C (2016). Prevalence of substance use disorders in psychiatric patients: a nationwide Danish population-based study. Soc Psychiatry Psychiatr Epidemiol, 51, 129140.Google Scholar
Ulrichsen, J, Nielsen, MK, Ulrichsen, M (2010). Disulfiram in severe alcoholism – an open controlled study. Nord J Psychiatry, 64, 356362.Google Scholar
Umhau, JC, Momenan, R, Schwandt, ML, et al. (2010). Effect of acamprosate on magnetic resonance spectroscopy measures of central glutamate in detoxified alcohol-dependent individuals: a randomized controlled experimental medicine study. Arch Gen Psychiatry, 67, 10691077.Google Scholar
Victorri-Vigneau, C, Dailly, E, Veyrac, G, Jolliet, P (2007). Evidence of zolpidem abuse and dependence: results of the French Centre for Evaluation and Information on Pharmacodependence (CEIP) network survey. Br J Clin Pharmacol, 64, 198209.Google Scholar
Weaver, T, Madden, P, Charles, V, et al. (2003). Comorbidity of substance misuse and mental illness in community mental health and substance misuse services. Br J Psychiatry, 183, 304313.Google Scholar
Wesson, DR, Ling, W (2003). The Clinical Opiate Withdrawal Scale (COWS). J Psychoactive Drugs, 35, 253259.Google Scholar
Wilde, MI, Wagstaff, AJ (1997). Acamprosate. Drugs, 53, 10381053.Google Scholar
World Health Organization (1992). ICD-10: International Statistical Classification of Diseases and Related Health Problems. Geneva: World Health Organization.Google Scholar
World Health Organization (2014). Global status report on alcohol and health 2014. Available at: www.who.int/substance_abuse/publications/global_alcohol_report/en/ (last accessed 11.8.19).Google Scholar

References

Aaronson, ST, Carpenter, LL, Conway, CR, et al. (2012). Vagus nerve stimulation therapy randomized to different amounts of electrical charge for treatment-resistant depression: acute and chronic effects. Brain Stimul, 6, 631640.Google Scholar
Aaronson, ST, Sears, P, Ruvuna, F, et al. (2017). A 5-year observational study of patients with treatment-resistant depression treated with vagus nerve stimulation or treatment as usual: comparison of response, remission, and suicidality. Am J Psychiatry, 174, 640648.Google Scholar
Abelson, JL, Curtis, GC, Sagher, O, et al. (2005). Deep brain stimulation for refractory obsessive-compulsive disorder. Biol Psychiatry, 57, 510516.Google Scholar
Adderley, DJ, Hamilton, M (1953). Use of succinylcholine in ECT. BMJ, 1, 195197.Google Scholar
Aleman, A, Sommer, IE, Kahn, RS (2007). Efficacy of slow repetitive transcranial magnetic stimulation in the treatment of resistant auditory hallucinations in schizophrenia: a meta-analysis. J Clin Psychiatry, 68, 416421.Google Scholar
American Psychiatric Association (2002). Practice Guideline for the Treatment of Patients with Bipolar Disorder, 2nd ed. Arlington, VA: American Psychiatric Association.Google Scholar
American Psychiatric Association (2010). Practice Guideline for the Treatment of Patients with Major Depressive Disorder, 3rd ed. Washington, DC: American Psychiatric Association. Available at: http://dx.doi.org/10.1176/appi.books.9780890423387.654001 (last accessed 31.8.19).Google Scholar
Arfai, E, Theano, G, Montagu, JD, Robin, AA (1970). A controlled study of polarization in depression. Br J Psychiatry, 116, 433434.Google Scholar
Baeken, C, De Raedt, R (2011). Neurobiological mechanisms of repetitive transcranial magnetic stimulation on the underlying neuro circuitry in unipolar depression. Dialogues Clin Neurosci, 13, 139145.Google Scholar
Bailey, P, Bremer, F (1938). A sensory cortical representation of the vagus nerve (with a note on the effects of low blood pressure on the cortical electrogram). J Neurophysiol, 1, 405412.Google Scholar
Bajbouj, M, Merkl, A, Schlaepfer, TE, et al. (2010). Two-year outcome of vagus nerve stimulation in treatment-resistant depression. J Clin Psychopharmacol, 30, 273281.Google Scholar
Barker, AT, Jalinous, R, Freeston, IL (1985). Non-invasive magnetic stimulation of human motor cortex. Lancet, 325, 11061107. http://dx.doi.org/http://dx.doi.org/10.1016/S0140-6736(85)92413-4.Google Scholar
Ben-Menachem, E, Manon-Espaillat, R, Ristanovic, R, et al. (1994). Vagus nerve stimulation for treatment of partial seizures: 1. A controlled study of effect on seizures. Epilepsia, 35, 616626.Google Scholar
Benbow, SM, White, J (2013). Safe ECT practice in people with a physical illness. In Waite, J, Easton, A, eds., The ECT Handbook, 3rd ed. London: Royal College of Psychiatrists, pp. 184190.Google Scholar
Berlim, MT, Van den Eynde, F, Daskalakis, ZJ (2013). Efficacy and acceptability of high frequency repetitive transcranial magnetic stimulation (rTMS) versus electroconvulsive therapy (ECT) for major depression: a systematic review and meta-analysis of randomized trials. Depress Anxiety, 30, 614623.Google Scholar
Braga, RJ, Petrides, G (2007). Somatic therapies for treatment-resistant psychiatric disorders. Braz J Pschiatry, 29(Suppl 2), S77S84.Google Scholar
Brakemeier, EL, Merkl, A, Wilbertz, G, et al. (2014). Cognitive-behavioral therapy as continuation treatment to sustain response after electroconvulsive therapy in depression: a randomized controlled trial. Biol Psychiatry, 76, 194202.Google Scholar
Brunoni, AR, Moffa, AH, Fregni, F, et al. (2016). Transcranial direct current stimulation for acute major depressive episodes: meta-analysis of individual patient data. Br J Psychiatry, 208, 522531.Google Scholar
Brus, O, Nordanskog, P, Båve, U, et al. (2017). Subjective memory immediately following electroconvulsive therapy. J ECT, 33, 96103.Google Scholar
Buley, N, Copland, E, Hodge, S (2017). ECT Minimum Dataset 2016–17: Activity Data Report – England, Wales, Northern Ireland & Republic of Ireland. London: Royal College of Psychiatrists.Google Scholar
Burke, MJ, Husain, MM (2006). Concomitant use of vagus nerve stimulation and electroconvulsive therapy for treatment-resistant depression. J ECT, 22, 218222.Google Scholar
Chervyakov, AV, Chernyavsky, AY, Sinitsyn, DO, Piradov, MA (2015). Possible mechanisms underlying the therapeutic effects of transcranial magnetic stimulation. Front Hum Neurosci, 9, 303.Google Scholar
Chiken, S, Nambu, A (2016). Mechanism of deep brain stimulation: inhibition, excitation, or disruption? Neuroscientist, 22, 313322.Google Scholar
Christmas, D, Steele, JD, Tolomeo, S, Eljamel, MS, Matthews, K (2013). Vagus nerve stimulation for chronic major depressive disorder: 12-month outcomes in highly treatment-refractory patients. J Affect Disord, 150, 12211225.Google Scholar
Cleare, A, Pariante, CM, Young, AH, et al. (2015). Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 2008 British Association for Psychopharmacology guidelines. J Psychopharmacol, 29, 459525.Google Scholar
de Vreede, IM, Burger, H, van Vliet, IM (2005). Prediction of response to ECT with routinely collected data in major depression. J Affect Disord, 86, 323327.Google Scholar
DeGiorgio, C, Heck, C, Bunch, S, et al. (2005). Vagus nerve stimulation for epilepsy: randomized comparison of three stimulation paradigms. Neurology, 65, 317319.Google Scholar
Deng, ZD, Hardesty, DE, Lisanby, SH, Peterchev, AV (2010). Electroconvulsive therapy in the presence of deep brain stimulation implants: electric field effects. Engineering in Medicine and Biology Society (EMBC), 2010 Annual International Conference of the IEEE 2010, 2049–2052.Google Scholar
Denys, D, Mantione, M, Figee, M, et al. (2010). Deep brain stimulation of the nucleus accumbens for treatment-refractory obsessive-compulsive disorder. Arch Gen Psychiatry, 67, 10611068.Google Scholar
Dlabac-de Lange, JJ, Bais, L, van Es, FD, et al. (2015). Efficacy of bilateral repetitive transcranial magnetic stimulation for negative symptoms of schizophrenia: results of a multicenter double-blind randomized controlled trial. Psychol Med, 45, 12631275.Google Scholar
Dollfus, S, Lecardeur, L, Morello, R, Etard, O (2016). Placebo response in repetitive transcranial magnetic stimulation trials of treatment of auditory hallucinations in schizophrenia: a meta-analysis. Schizophr Bull, 42, 301308.Google Scholar
Enomoto, S, Shimizu, K, Nibuya, M, et al. (2017). Activated brain-derived neurotrophic factor/TrkB signaling in rat dorsal and ventral hippocampi following 10-day electroconvulsive seizure treatment. Neurosci Lett, 660, 4550.Google Scholar
Erickson, JM, Carty, J (2015). Safe and effective electroconvulsive therapy using multiple parameters over 5 years in a patient with deep brain stimulator. J ECT, 31, 278279.Google Scholar
EUnetHTA (2017). Repetitive Transcranial Magnetic Stimulation for Treatment-Resistant Major Depression. Project ID: WP4-ACB-CA-5. Diemen: European Network for Health Technology Assessment.Google Scholar
FDA (2011). FDA Executive Summary. Prepared for the January 27–28, 2011 meeting of the Neurological Devices Panel: Meeting to Discuss the Classification of Electroconvulsive Therapy Devices (ECT). Silver Spring, MD: US Food and Drug Administration.Google Scholar
Fox, MD, Buckner, RL, White, MP, Greicius, MD, Pascual-Leone, A (2012). Efficacy of transcranial magnetic stimulation targets for depression is related to intrinsic functional connectivity with the subgenual cingulate. Biol Psychiatry, 72, 595603.Google Scholar
Fraser, LM, O’Carroll, RE, Ebmeier, KP (2008). The effect of electroconvulsive therapy on autobiographical memory: a systematic review. J ECT, 24, 1017.Google Scholar
Frye, RE, Rotenberg, A, Ousley, M, Pascual-Leone, A (2008). Transcranial magnetic stimulation in child neurology: current and future directions. J Child Neurol, 23, 7996.Google Scholar
Gbyl, K, Videbech, P (2018). Electroconvulsive therapy increases brain volume in major depression: a systematic review and meta-analysis. Acta Psychiatr Scand, 138, 180195.Google Scholar
George, MS, Rush, AJ, Marangell, LB, et al. (2005). A one-year comparison of vagus nerve stimulation with treatment as usual for treatment-resistant depression. Biol Psychiatry, 58, 364373.Google Scholar
Goodman, WK, Foote, KD, Greenberg, BD, et al. (2010). Deep brain stimulation for intractable obsessive compulsive disorder: pilot study using a blinded, staggered-onset design. Biol Psychiatry, 67, 535542.Google Scholar
Goodwin, GM, Haddad, PM, Ferrier, IN, et al. (2016). Evidence-based guidelines for treating bipolar disorder: revised third edition recommendations from the British Association for Psychopharmacology. J Psychopharmacol, 30, 495553.Google Scholar
Goyal, N, Nizamie, SH, Desarkar, P (2007). Efficacy of adjuvant high frequency repetitive transcranial magnetic stimulation on negative and positive symptoms of schizophrenia: preliminary results of a double-blind sham-controlled study. J Neuropsychiatry Clin Neurosci, 19, 464467.Google Scholar
Gupta, N, Avasthi, A, Kulhara, P (2000). Clinical variables as predictors of response to electroconvulsive therapy in endogenous depression. Indian J Psychiatry, 42, 6065.Google Scholar
Hawkins, JM, Archer, KJ, Strakowski, SM, Keck, PE (1995). Somatic treatment of catatonia. Int J Psychiatry Med, 25, 345369.Google Scholar
Hazari, H, Christmas, D, Matthews, K (2013). The clinical utility of different quantitative methods for measuring treatment resistance in major depression. J Affect Disord, 150, 231236.Google Scholar
Health Quality Ontario (2016). Repetitive transcranial magnetic stimulation for treatment-resistant depression: a systematic review and meta-analysis of randomized controlled trials. Ont Health Technol Assess Ser, 16, 166.Google Scholar
Hein, E, Nowak, M, Kiess, O, et al. (2013). Auricular transcutaneous electrical nerve stimulation in depressed patients: a randomized controlled pilot study. J Neural Transm (Vienna), 120, 821827.Google Scholar
Hızlı Sayar, G, Ozten, E, Tufan, E, et al. (2014). Transcranial magnetic stimulation during pregnancy. Arch Womens Ment Health, 17, 311315.Google Scholar
Ho, K-A, Taylor, JL, Loo, CK (2015). Comparison of the effects of transcranial random noise stimulation and transcranial direct current stimulation on motor cortical excitability. J ECT, 31, 6772.Google Scholar
Holtzheimer, PE, Husain, MM, Lisanby, SH, et al. (2017). Subcallosal cingulate deep brain stimulation for treatment-resistant depression: a multisite, randomised, sham-controlled trial. Lancet Psychiatry, 4, 839849. http://dx.doi.org/10.1016/S2215-0366(17)30371-1.Google Scholar
Horvath, JC, Forte, JD, Carter, O (2015). Quantitative review finds no evidence of cognitive effects in healthy populations from single-session transcranial direct current stimulation (tDCS). Brain Stimul, 8, 535550.Google Scholar
International Neuromodulation Society (2016). Welcome to the International Neuromodulation Society. Available at: www.neuromodulation.com (last accessed 21.12.16).Google Scholar
Jelovac, A, Kolshus, E, McLoughlin, DM (2013). Relapse following successful electroconvulsive therapy for major depression: a meta-analysis. Neuropsychopharmacology, 38, 24672474.Google Scholar
Jelovac, A, O’Connor, S, McCarron, S, McLoughlin, DM (2016). Autobiographical memory specificity in major depression treated with electroconvulsive therapy. J ECT, 32, 3843.Google Scholar
Kellner, CH, Knapp, RG, Petrides, G, et al. (2006). Continuation electroconvulsive therapy vs pharmacotherapy for relapse prevention in major depression: a multisite study from the Consortium for Research in Electroconvulsive Therapy (CORE). Arch Gen Psychiatry, 63, 13371344.Google Scholar
Kellner, CH, Knapp, R, Husain, MM, et al. (2010). Bifrontal, bitemporal and right unilateral electrode placement in ECT: randomised trial. Br J Psychiatry, 196, 226234.Google Scholar
Kindler, S, Shapira, B, Hadjez, J, et al. (1991). Factors influencing response to bilateral electroconvulsive therapy in major depression. Convuls Ther, 7, 245254.Google Scholar
Krystal, AD, Coffey, CE (1997). Neuropsychiatric considerations in the use of electroconvulsive therapy. J Neuropsychiatry Clin Neurosci, 9, 283292.Google Scholar
Leroy, A, Naudet, F, Vaiva, G, et al. (2018). Is electroconvulsive therapy an evidence-based treatment for catatonia? A systematic review and meta-analysis. Eur Arch Psychiatry Clin Neurosci, 268, 675687.Google Scholar
Limousin, P, Pollak, P, Benazzouz, A, et al. (1995). Effect of parkinsonian signs and symptoms of bilateral subthalamic nucleus stimulation. Lancet, 345, 9195.Google Scholar
Lisanby, SH, Luber, B, Schlaepfer, TE, Sackeim, HA (2003). Safety and feasibility of magnetic seizure therapy (MST) in major depression: randomized within-subject comparison with electroconvulsive therapy. Neuropsychopharmacology, 28, 18521865.Google Scholar
Loo, CK, Mahon, M, Katalinic, N, Lyndon, B, Hadzi-Pavlovic, D (2011). Predictors of response to ultrabrief right unilateral electroconvulsive therapy. J Affect Disord, 130, 192197.Google Scholar
Madsen, TM, Treschow, A, Bengzon, J, et al. (2000). Increased neurogenesis in a model of electroconvulsive therapy. Biol Psychiatry, 47, 10431049.Google Scholar
Mallet, L, Polosan, M, Jaafari, N, et al. (2008). Subthalamic nucleus stimulation in severe obsessive-compulsive disorder. N Engl J Med, 359, 21212134.Google Scholar
Marangell, LB, Rush, AJ, George, MS, et al. (2002). Vagus nerve stimulation (VNS) for major depressive episodes: one year outcomes. Biol Psychiatry, 51, 280287.Google Scholar
Martin, JLR, Martín-Sánchez, E (2012). Systematic review and meta-analysis of vagus nerve stimulation in the treatment of depression: variable results based on study designs. Eur Psychiatry, 27, 147155.Google Scholar
Mayberg, HS (2009). Targeted electrode-based modulation of neural circuits for depression. J Clin Invest, 119, 717725.Google Scholar
Mayberg, HS, Lozano, AM, Voon, V, et al. (2005). Deep brain stimulation for treatment-resistant depression. Neuron, 45, 651660.Google Scholar
McClelland, J, Kekic, M, Bozhilova, N, et al. (2016). A randomised controlled trial of neuronavigated repetitive transcranial magnetic stimulation (rTMS) in anorexia nervosa. PLoS One , 11, e0148606.Google Scholar
Medda, P, Mauri, M, Toni, C, et al. (2014). Predictors of remission in 208 drug-resistant depressive patients treated with electroconvulsive therapy. J ECT, 30, 292297.Google Scholar
Milev, RV, Giacobbe, P, Kennedy, SH, et al. (2016). Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 Clinical Guidelines for the Management of Adults with Major Depressive Disorder: Section 4. Neurostimulation Treatments. Can J Psychiatry, 61, 561575.Google Scholar
Mu, Q, Bohning, DE, Nahas, Z, et al. (2004). Acute vagus nerve stimulation using different pulse widths produces varying brain effects. Biol Psychiatry, 55, 816825.Google Scholar
Munk-Olsen, T, Laursen, TM, Videbech, P, Mortensen, PB, Rosenberg, R (2007). All-cause mortality among recipients of electroconvulsive therapy: register-based cohort study. Br J Psychiatry, 190, 435459.Google Scholar
Nahas, Z, Marangell, LB, Husain, MM, et al. (2005). Two-year outcome of vagus nerve stimulation (VNS) for treatment of major depressive episodes. J Clin Psychiatry, 66, 10971104.Google Scholar
Nardone, R, Tezzon, F, Höller, Y, et al. (2014). Transcranial magnetic stimulation (TMS)/repetitive TMS in mild cognitive impairment and Alzheimer’s disease. Acta Neurol Scand, 129, 351366.Google Scholar
National Institute for Clinical Excellence (2003). Guidance on the use of electroconvulsive therapy. Technical appraisal guidance [TA59]. London: National Collaborating Centre for Mental Health. Available at: www.nice.org.uk/Guidance/TA59 (last accessed 31.8.19).Google Scholar
National Institute for Health and Care Excellence (NICE) (2009). Vagus nerve stimulation for treatment-resistant depression. Interventional procedures guidance [IPG330]. London: National Collaborating Centre for Mental Health.Google Scholar
National Institute for Health and Care Excellence (NICE) (2015). Repetitive transcranial magnetic stimulation for depression. Interventional procedures guidance [IPG542]. London: National Collaborating Centre for Mental Health. Available at: http://nice.org.uk/guidance/ipg542 (last accessed 31.8.19).Google Scholar
Navarro, V, Gasto, C, Torres, X, et al. (2008). Continuation/maintenance treatment with nortriptyline versus combined nortriptyline and ECT in late-life psychotic depression: a two-year randomized study. Am J Geriatr Psychiatry, 16, 498505.Google Scholar
Ng, C, Schweitzer, I, Alexopoulos, P, et al. (2000). Efficacy and cognitive effects of right unilateral electroconvulsive therapy. Journal of ECT, 16, 370379.Google Scholar
Nordenskjöld, A, von Knorring, L, Engström, I (2012). Predictors of the short-term responder rate of electroconvulsive therapy in depressive disorders – a population based study. BMC Psychiatry, 12, 115.Google Scholar
Nutt, DJ, Gleiter, CH, Glue, P (1989). Neuropharmacological aspects of ECT: in search of the primary mechanism of action. Convul Ther, 5, 250260.Google Scholar
Nuttin, B, Cosyns, P, Demeulemeester, H, Gybels, J, Meyerson, B (1999). Electrical stimulation in anterior limbs of internal capsules in patients with obsessive-compulsive disorder. Lancet, 354, 1526.Google Scholar
Oltedal, L, Narr, KL, Abbott, C, et al. (2018). Volume of the human hippocampus and clinical response following electroconvulsive therapy. Biol Psychiatry, 84, 574581.Google Scholar
Palm, U, Hasan, A, Strube, W, Padberg, F (2016). tDCS for the treatment of depression: a comprehensive review. Eur Arch Psychiatry Clin Neurosci, 266, 681694.Google Scholar
Pande, AC, Krugler, T, Haskett, RF, Greden, JF, Grunhaus, LJ (1988). Predictors of response to electroconvulsive therapy in major depressive disorder. Biol Psychiatry, 24, 9193.Google Scholar
Penry, JK, Dean, JC (1990). Prevention of intractable partial seizures by intermittent vagal stimulation in humans: preliminary results. Epilepsia, 31(Suppl 2), S40S43.Google Scholar
Perrin, JS, Merz, S, Bennett, DM, et al. (2012). Electroconvulsive therapy reduces frontal cortical connectivity in severe depressive disorder. Proc Natl Acad Sci U S A, 109, 54645468.Google Scholar
Philip, NS, Barredo, J, van ’t Wout-Frank, M, et al. (2018). Network mechanisms of clinical response to transcranial magnetic stimulation in posttraumatic stress disorder and major depressive disorder. Biol Psychiatry, 83, 263272.Google Scholar
Pinna, M, Manchia, M, Oppo, R, et al. (2018). Clinical and biological predictors of response to electroconvulsive therapy (ECT): a review. Neurosci Lett, 669, 3242.Google Scholar
Prudic, J, Sackeim, HA, Devanand, DP (1990). Medication resistance and clinical response to electroconvulsive therapy. Psychiatry Res, 31, 287296.Google Scholar
RCPsych Committee on ECT and Related Treatments (2017). Statement on Neurosurgery for Mental Disorder (NMD), also known as Psychiatric Neurosurgery (Position statement CERT 05/17). London: Royal College of Psychiatrists.Google Scholar
Ren, J, Li, H, Palaniyappan, L, et al. (2014). Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depression: a systematic review and meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry, 51, 181189.Google Scholar
Rocha, RB, Dondossola, ER, Grande, AJ, et al. (2016). Increased BDNF levels after electroconvulsive therapy in patients with major depressive disorder: a meta-analysis study. J Psychiatr Res, 83, 4753.Google Scholar
Rong, P, Liu, J, Wang, L, et al. (2016). Effect of transcutaneous auricular vagus nerve stimulation on major depressive disorder: a nonrandomized controlled pilot study. J Affect Disord, 195, 172179.Google Scholar
Rossi, S, Hallett, M, Rossini, PM, Pascual-Leone, A; Safety of TMS Consensus Group (2009). Safety, ethical considerations, and application guidelines for the use of transcranial magnetic stimulation in clinical practice and research. Clin Neurophysiology, 120, 20082039.Google Scholar
Rush, AJ, George, MS, Sackeim, HA, et al. (2000). Vagus nerve stimulation (VNS) for treatment-resistant depressions: a multicenter study. Biol Psychiatry, 47, 276286.Google Scholar
Rush, AJ, Marangell, LB, Sackeim, HA, et al. (2005a). Vagus nerve stimulation for treatment-resistant depression: a randomized, controlled acute phase trial. Biol Psychiatry, 58, 347354.Google Scholar
Rush, AJ, Sackei, HA, Marangell, LB, et al. (2005b). Effects of 12 months of vagus nerve stimulation in treatment-resistant depression: a naturalistic study. Biol Psychiatry, 58, 355363.Google Scholar
Sackeim, HA, Prudic, J, Devanand, DP, et al. (2000). A prospective, randomized, double-blind comparison of bilateral and right unilateral electroconvulsive therapy at different stimulus intensities. Arch Gen Psychiatry, 57, 425434.Google Scholar
Sackeim, HA, Haskett, RF, Mulsant, BH, et al. (2001). Continuation pharmacotherapy in the prevention of relapse following electroconvulsive therapy: a randomized controlled trial. JAMA, 285, 12991307.Google Scholar
Saleh, C, Fontaine, D (2015). Deep brain stimulation for psychiatric diseases: what are the risks? Curr Psychiatry Rep, 17, 114.Google Scholar
SEAN (2016). SEAN Annual Report 2016. A Summary of ECT in Scotland for 2015. Edinburgh: Scottish ECT Audit Network.Google Scholar
SEAN (2017). SEAN Annual Report 2017. A Summary of ECT in Scotland for 2016. Edinburgh: Scottish ECT Audit Network.Google Scholar
Semkovska, M, McLoughlin, DM (2010). Objective cognitive performance associated with electroconvulsive therapy for depression: a systematic review and meta-analysis. Biol Psychiatry, 68, 568577.Google Scholar
Sheline, YI, Price, JL, Yan, Z, Mintun, MA (2010). Resting-state functional MRI in depression unmasks increased connectivity between networks via the dorsal nexus. Proc Natl Acad Sci U S A, 107, 1102011025.Google Scholar
Shiwach, RS, Reid, WH, Carmody, TJ (2001). An analysis of reported deaths following electroconvulsive therapy in Texas, 1993–1998. Psychiatr Serv, 52, 10951097.Google Scholar
Straasø, B, Lauritzen, L, Lunde, M, et al. (2014). Dose-remission of pulsating electromagnetic fields as augmentation in therapy-resistant depression: a randomized, double-blind controlled study. Acta Neuropsychiatr, 26, 272279.Google Scholar
Tharyan, P, Adams, CE (2005). Electroconvulsive therapy for schizophrenia. Cochrane Database Syst Rev, (2), CD000076. doi:10.1002/14651858.CD000076.pub2.Google Scholar
Tor, PC, Bautovich, A, Wang, MJ, et al. (2015). A systematic review and meta-analysis of brief versus ultrabrief right unilateral electroconvulsive therapy for depression. J Clin Psychiatry, 76, e1092e1098.Google Scholar
TrevizoL, AP, Shiozawa, P, Cook, IA, et al. (2016). Transcranial magnetic stimulation for obsessive-compulsive disorder: an updated systematic review and meta-analysis. J ECT, 32, 262266.Google Scholar
Tyagi, H, Zrinzo, L, Akram, H, et al. (2017). A randomised controlled trial of deep brain stimulation in obsessive compulsive disorder: a comparison of ventral capsule/ventral striatum and subthalamic nucleus targets. J Neurol Neurosurg Psychiatry, 88, A8A9.Google Scholar
UK ECT Review Group (2003). Systematic Review of the Efficacy and Safety of Electroconvulsive Therapy. London: Department of Health.Google Scholar
van Schaik, AM, Comijs, HC, Sonnenberg, CM, et al. (2012). Efficacy and safety of continuation and maintenance electroconvulsive therapy in depressed elderly patients: a systematic review. Am J Geriatr Psychiatry, 20, 517.Google Scholar
Vila-Rodriguez, F, McGirr, A, Tham, J, Hadjipavlou, G, Honey, CR (2014). Electroconvulsive therapy in patients with deep brain stimulators. J ECT, 30, e16e18.Google Scholar
Watts, BV, Groft, A, Bagian, JP, Mills, PD (2011). An examination of mortality and other adverse events related to electroconvulsive therapy using a national adverse event report system. J ECT, 27, 105108.Google Scholar
Xia, G, Gajwani, P, Muzina, DJ, et al. (2008). Treatment-emergent mania in unipolar and bipolar depression: focus on repetitive transcranial magnetic stimulation. Int J Neuropsychopharmacology, 11, 119130.Google Scholar
Zhou, D-D, Wang, W, Wang, G-M, Li, D-Q, Kuang, L (2017). An updated meta-analysis: short-term therapeutic effects of repeated transcranial magnetic stimulation in treating obsessive-compulsive disorder. J Affect Disord, 215, 187196.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×